Movatterモバイル変換


[0]ホーム

URL:


WO2024236156A1 - Anti-cathepsin-d antibodies - Google Patents

Anti-cathepsin-d antibodies
Download PDF

Info

Publication number
WO2024236156A1
WO2024236156A1PCT/EP2024/063625EP2024063625WWO2024236156A1WO 2024236156 A1WO2024236156 A1WO 2024236156A1EP 2024063625 WEP2024063625 WEP 2024063625WWO 2024236156 A1WO2024236156 A1WO 2024236156A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
cells
cell
cathepsin
tumor
Prior art date
Application number
PCT/EP2024/063625
Other languages
French (fr)
Inventor
Emmanuelle LIAUDET-COOPMAN
Thierry Chardes
Thimothée DAVID
Pénélope DESROYS DU ROURE
Original Assignee
Institut National de la Santé et de la Recherche Médicale
Institut Régional Du Cancer De Montpellier
Université De Montpellier
Centre National De La Recherche Scientifique
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut National de la Santé et de la Recherche Médicale, Institut Régional Du Cancer De Montpellier, Université De Montpellier, Centre National De La Recherche ScientifiquefiledCriticalInstitut National de la Santé et de la Recherche Médicale
Publication of WO2024236156A1publicationCriticalpatent/WO2024236156A1/en

Links

Classifications

Definitions

Landscapes

Abstract

The inventors generated a new anti-Cath-D antibody (F1M1) which is able to reduce tumor growth without apparent toxicity in strongly immune-infiltrated Cath-D-secreting basal-like TNBC cell lines. F1M1 antibody prevented the recruitment of immunosuppressive M2- polarized tumor-associated macrophages (TAMs), and induced natural killer cell activation in tumors, F1M1 also enhanced the activation of anti -tumor M1 -polarized TAMs, the recruitment and maturation of conventional cDC1 dendritic cells in tumors to promote antigen presentation, and reduced the expression of exhaustion markers on CD4+ and CD8+ T cells in tumors and draining lymph nodes. Interestingly, the antibody F1M1 exhibit also a better affinity to Cath-D than the antibody F1 previously generated by the inventors. The inventors also generated a novel Fc-optimized F1M 1-Fc+ human antibody that promotes ADCC induction on both cancer cells and CAF, improves antitumor potency, and triggers NK cell recruitment, activation and cytotoxic activity in tumors. F1M1-Fc+ F1M1-Fc+ inhibited growth of MDA-MB-231 and SUM159 TNBC cell xenografts, and of two TNBC-PDX (one resistant to neoadjuvant chemotherapy) without apparent toxicity. Moreover, this F1M1-Fc+ improves paclitaxel and enzalutamide therapeutic efficacy in combination. Thus, the present invention relates to anti- cathepsin-D antibodies and their use in the treatment of cancers, particularly of triple negative breast cancer.

Description

ANTI-CATH-D ANTIBODIES FIELD OF THE INVENTION: The present invention relates to cancer field. More particularly, the invention relates to anti-cathepsin-D antibodies and their uses in the treatment of cancers, particularly of triple negative breast cancer. BACKGROUND OF THE INVENTION: Triple-negative breast cancers (TNBC; 15% of all breast cancers, BC) are defined by the lack of oestrogen receptor, progesterone receptor and HER2 expression/amplification (1). The prognosis of patients with TNBC is poor, mainly due to the disease heterogeneity and lack of targeted therapies. Immunotherapies that exploit the immune system to promote durable tumor regression and prolong survival are an interesting strategy for TNBC, which is classified as an immunogenic BC subtype due to its high tumor mutational burden and presence of immune cell infiltrates (2,3). The median tumor mutational burden, which is the main source of neoantigens to induce anti-tumor immunity, is higher in TNBC than in other BC subtypes, including the HER2-amplified BC subtype (3). In metastatic TNBC, immune checkpoint inhibitors (ICI) (anti-PD-1 and anti-PD-L1 antibodies) and chemotherapy have improved survival in patients with PD-L1-expressing tumors (4). ICI are also a relevant option for patients with HER2-positive BC in metastatic, neoadjuvant and adjuvant settings (5). In BC, tumor-associated macrophages (TAMs) are the most abundant inflammatory cells. Typically, they are M2-polarized cells with suppressive capacity (6) linked to their enzymatic activities and anti-inflammatory cytokine production. In TNBC, high M2-polarized TAM levels are associated with poorer outcome (7), whereas tumor-infiltrating lymphocytes are associated with improved disease-free and overall survival rates (8). To exhibit anti-tumor function, lymphocytes must: i) be primed and activated by antigen-presenting cells (APCs), such as dendritic cells (DCs) and M1-polarized anti-tumor TAMs, and ii) accumulate in the tumor microenvironment, while avoiding the escape mechanisms induced by this immunosuppressive microenvironment. Antibody-based therapies can modulate the recruitment, activation and immunosuppression of immune cells in the tumor microenvironment (1,2). Importantly, recent studies and clinical trials highlighted that NK cell-based immunotherapy can awake the innate anticancer response, particularly against tumor metastases (9-11), and sustain breast cancer dormancy (12). Hence, different immunotherapeutic strategies are tested to improve the efficacy of antibody-induced NK cell-mediated antitumor activity. Glyco-engineering and protein-engineering of the Fc region of tumor-targeting antibodies have been exploited to modulate their interaction with activating or inhibitory members of the FcγR family (13). Afucosylation and selected amino acid substitutions increase the affinity for FcγRs, thus enhancing NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) (13), a major mechanism contributing to the therapeutic efficacy of antibodies (14). Many clinically- relevant anticancer antibodies, such as rituximab, cetuximab and trastuzumab, induce NK cell- mediated ADCC in vitro (11, 13). In the tumor microenvironment (stroma), NK cells bind to the Fc of antibody-labeled tumor cells, predominantly through a single activating FcγR (FcγRIIIA, also named CD16a), a low-affinity receptor for IgG (15). Binding of IgG to CD16a can activate NK cells and stimulate the release of lytic granules that contain molecules, such as perforin and granzymes (16,17) ultimately leading to the target cell lysis. FcγR ligation also affects NK cell survival and proliferation (18), and induces the release of cytokines and chemokines that promote the recruitment and activation of tumor-infiltrating immune cells (15). The aspartic protease cathepsin D (Cath-D), a poor prognosis marker in BC (19) including TNBC (20), is overproduced by BC cells and hypersecreted in the tumor microenvironment (21). In TNBC, Cath-D is a tumor cell-associated extracellular biomarker (22) with tumor-promoting activity (23-18)), and a potent target for antibody-based therapy (22). We previously showed that F1, a fully human anti-Cath-D IgG1, activates natural killer (NK) cells and prevents the tumor recruitment of M2-polarized TAMs and myeloid-derived immunosuppressive populations in immunodeficient Foxn1nu nude mice (22). Therefore, due to its immunomodulatory activity, F1 could represent a promising option for patients with chemotherapy- and/or ICI-resistant TNBC. However, the overall impact of anti-Cath-D antibody-based therapy on immune cell recruitment in fully immunocompetent mouse models remained unknown, particularly in function of the tumor immune microenvironment. Here, the inventor investigated the antitumor efficacy and immunomodulatory activity of anti-Cath-D antibodies in a fully immunocompetent mouse model of highly immune cell infiltrated TNBC. They also explored whether targeted therapy with Fc-engineered human anti-cath-D antibodies triggers ADCC, and the relevance of combination therapies with anti-cath-D antibodies in TNBC. SUMMARY OF THE INVENTION: The present invention shows that anti-Cath-D antibodies trigger the antitumor innate and adaptive immune responses in preclinical immunocompetent mouse models of breast cancer (TNBC). Anti-Cath-D antibodies represent a promising immunotherapy for patients with immunogenic TNBC. The present invention also shows that Cath-D is a tumor microenvironment antigen eligible for Fc-engineered antibody targeted therapy to trigger ADCC. The Fc-optimized F1M1-Fc+ antibody (derived from F1M1 by introducing the S239D, H268F, S324T, and I332E mutations to enhance the affinity for CD16a) promotes ADCC induction, improves antitumor potency, and triggers NK cell recruitment, activation and cytotoxic activity in tumors. F1M1- Fc+ improves paclitaxel and enzalutamide therapeutic efficacy in combination. F1M1-Fc+ is a new potentially successful immunotherapy for TNBC that could be combined with conventional regimens, including chemotherapy or antiandrogens, and also ICIs. Thus, the present invention relates to an anti-cathepsin-D antibody which inhibits the tumor recruitment of immunosuppressive tumor-associated macrophages M2 and myeloid- derived suppressor cells and which activate NK cells in the stromal tumor microenvironment. The present invention also relates to an Fc-engineered anti-cathepsin-D antibody that triggers ADCC induction, NK cell recruitment, activation, and cytotoxic activity and improved antitumor activity in TNBC. The present invention also related to method for treating hyperproliferative disorders, comprising administering to said subject an effective amount of the antibody of the invention. In particular, the invention is defined by the claims. DETAILED DESCRIPTION OF THE INVENTION: The inventors generated a new anti-Cath-D antibody (F1M1) which is able to reduce tumor growth without apparent toxicity in C57BL/6 mice harboring grafts of mouse E0771 cells (a strongly immune-infiltrated Cath-D-secreting basal-like TNBC cell line). In the E0771 model, F1M1 antibody prevented the recruitment of immunosuppressive M2-polarized tumor- associated macrophages (TAMs), and induced natural killer cell activation in tumors, as shown by 17-color flow cytometry immunophenotyping. F1M1 also enhanced the activation of anti- tumor M1-polarized TAMs, and the recruitment and maturation of conventional cDC1 dendritic cells in tumors to promote antigen presentation. Lastly, F1M1 reduced the expression of exhaustion markers on CD4+ and CD8+ T cells in tumors and draining lymph nodes. The inventors also applied Fc-engineering to F1M1, a novel fully human anti-Cath-D IgG1 antibody in which the variable regions were derived from F1, to generate three antibody variants carrying the wild-type (F1M1) or mutated Fc region to enhance (F1M1-Fc+) or prevent (F1M1-Fc-) binding to CD16a on NK cells. F1M1-Fc+ induced ADCC against TNBC and stromal cells, such as breast CAF, by activating NK cells more efficiently than F1M1, whereas F1M1-Fc- was ineffective. In MDA-MB-231 cell xenografts (TNBC model), antitumor activity was higher with F1M1-Fc+ than with F1M1. F1M1-Fc- was the least effective, reflecting the importance of Fc-dependent mechanisms also in vivo. F1M1-Fc+ triggered NK cell recruitment, activation and cytotoxic activity in MDA-MB-231 cell xenografts. We confirmed F1M1-Fc+ potent antitumor activity in SUM159 (TNBC) cell xenografts and two TNBC PDXs. Finally, when combined with paclitaxel or enzalutamide, F1M1-Fc+ improved their therapeutic efficacy in TNBC, demonstrating its clinical relevance. Moreover, the new anti-Cath-D antibody F1M1 has been generated to abrogate Fab N- glycosylation that may lead to the production of glycoforms associated with the risk of immunogenic responses in patients. Interestingly, the antibody F1M1 exhibited also a better affinity for both human and mouse Cath-D than the antibody F1 previously generated by the inventors. Definitions As used herein, the term "Cathepsin-D" or “Cath-D” has its general meaning in the art and refers to lysosomal aspartic protease cathepsin-D. Cath-D is synthesized as the 52 kDa, catalytically inactive, precursor called pro-Cath-D. It is present in endosomes as an active 48 kDa single-chain intermediate that is subsequently converted in the lysosomes into the fully active mature protease, composed of a 34 kDa heavy and a 14 kDa light chains. The naturally occurring pro-Cath-D protein has an amino acid sequence shown in Genbank, Accession number NP_001900. Cath-D is well known for its roles in metastasis, angiogenesis, proliferation, and carcinogenesis in cancer. Cath-D is overproduced by breast cancer cells and hypersecreted in the tumor microenvironment (21). As used herein, the term "anti-Cathepsin-D antibody" refers to an antibody directed and having specificity for Cath-D. As used herein, the term "F1 antibody" refers to an antibody directed and having specificity for Cath-D previously generated by the inventors and described in WO2016/188911 and Ashraf et al.2019 (22). As used herein, the term "antibody" or "immunoglobulin" have the same meaning, and will be used equally in the present invention. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen. As such, the term antibody encompasses not only whole antibody molecules, but also antibody fragments as well as variants (including derivatives) of antibodies and antibody fragments. In natural antibodies, two heavy chains are linked to each other by disulfide bonds and each heavy chain is linked to a light chain by a disulfide bond. There are two types of light chain, lambda (1) and kappa (k). There are five main heavy chain classes (or isotypes) which determine the functional activity of an antibody molecule: IgM, IgD, IgG, IgA and IgE. Each chain contains distinct sequence domains. The light chain includes two domains, a variable domain (VL) and a constant domain (CL). The heavy chain includes four domains, a variable domain (VH) and three constant domains (CHI, CH2 and CH3, collectively referred to as CH). The variable regions of both light (VL) and heavy (VH) chains determine binding recognition and specificity to the antigen. The constant region domains of the light (CL) and heavy (CH) chains confer important biological properties such as antibody chain association, secretion, trans-placental mobility, complement binding, and binding to Fc receptors (FcR). The Fv fragment is the N-terminal part of the Fab fragment of an immunoglobulin and consists of the variable portions of one light chain and one heavy chain. The specificity of the antibody resides in the structural complementarity between the antibody combining site and the antigenic determinant. Antibody combining sites are made up of residues that are primarily from the hypervariable or complementarity determining regions (CDRs). Occasionally, residues from nonhypervariable or framework regions (FR) can participate to the antibody binding site or influence the overall domain structure and hence the combining site. Complementarity Determining Regions or CDRs refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native immunoglobulin binding site. The light and heavy chains of an immunoglobulin each have three CDRs, designated L-CDR1, L- CDR2, L- CDR3 and H-CDR1, H-CDR2, H-CDR3, respectively. An antigen-binding site, therefore, typically includes six CDRs, comprising the CDR set from each of a heavy and a light chain V region. Framework Regions (FRs) refer to amino acid sequences interposed between CDRs. Knowing the amino acid sequence of the desired sequence, one skilled in the art can readily produce said antibodies, by standard techniques for production of polypeptides. For instance, they can be synthesized using well-known solid phase method, preferably using a commercially available peptide synthesis apparatus (such as that made by Applied Biosystems, Foster City, California) and following the manufacturer’s instructions. Alternatively, antibodies of the invention can be synthesized by recombinant DNA techniques well-known in the art. For example, antibodies can be obtained as DNA expression products after incorporation of DNA sequences encoding the antibodies into expression vectors and introduction of such vectors into suitable eukaryotic or prokaryotic hosts that will express the desired antibodies, from which they can be later isolated using well-known techniques. As used herein, the term “Fc region” has its general meaning and refers to the region of an antibody begin in the hinge region just upstream of the papain cleavage site which defines IgG Fc chemically (about residue 216 EU numbering, taking the first residue of heavy chain constant region to be 114) and ending at its C-terminus. As used herein, the term “hinge region” includes the portion of a heavy chain molecule that joins the CH1 domain to the CH2 domain, e.g. from about position 216-230 according to the EU number system. This hinge region comprises approximately 25 residues and is flexible, thus allowing the two N-terminal antigen binding regions to move independently. Hinge regions can be subdivided into three distinct domains: upper, middle, and lower hinge domains. As used herein, the term “CH2 domain” includes the portion of a heavy chain molecule that extends, e.g., from about EU positions 231- 340. The CH2 domain is unique in that it is not closely paired with another domain. Rather, two N-linked branched carbohydrate chains are interposed between the two CH2 domains of an intact native IgG molecule. As used herein, the term “CH3 domain” includes the portion of a heavy chain molecule that extends approximately 110 residues from N-terminus of the CH2 domain, e.g., from about residue 341-446, EU numbering system). The CH3 domain typically forms the C-terminal portion of the antibody. In some immunoglobulins, however, additional domains may extend from CH3 domain to form the C-terminal portion of the molecule (e.g. the CH4 domain in the chain of IgM and the E chain of IgE). In the context of the invention, the amino acid residues of the antibody of the invention are numbered according to the IMGT numbering system. The IMGT unique numbering has been defined to compare the variable domains whatever the antigen receptor, the chain type, or the species (Lefranc M.-P., "Unique database numbering system for immunogenetic analysis" Immunology Today, 18, 509 (1997) ; Lefranc M.-P., "The IMGT unique numbering for Immunoglobulins, T cell receptors and Ig-like domains" The Immunologist, 7, 132-136 (1999).; Lefranc, M.-P., Pommié, C., Ruiz, M., Giudicelli, V., Foulquier, E., Truong, L., Thouvenin- Contet, V. and Lefranc, G., "IMGT unique numbering for immunoglobulin and T cell receptor variable domains and Ig superfamily V-like domains" Dev. Comp. Immunol., 27, 55-77 (2003).). In the IMGT unique numbering, the conserved amino acids always have the same position, for instance cysteine 23, tryptophan 41, hydrophobic amino acid 89, cysteine 104, phenylalanine or tryptophan 118. The IMGT unique numbering provides a standardized delimitation of the framework regions (FR1-IMGT: positions 1 to 26, FR2-IMGT: 39 to 55, FR3-IMGT: 66 to 104 and FR4-IMGT: 118 to 128) and of the complementarity determining regions: CDR1-IMGT: 27 to 38, CDR2-IMGT: 56 to 65 and CDR3-IMGT: 105 to 117. If the CDR3-IMGT length is less than 13 amino acids, gaps are created from the top of the loop, in the following order 111, 112, 110, 113, 109, 114, etc. If the CDR3-IMGT length is more than 13 amino acids, additional positions are created between positions 111 and 112 at the top of the CDR3-IMGT loop in the following order 112.1,111.1, 112.2, 111.2, 112.3, 111.3, etc. (http://www.imgt.org/IMGTScientificChart/Nomenclature/IMGT-FRCDRdefinition.html). As used herein the term "human antibody” is intended to include antibodies having variable and constant regions derived from human immunoglobulin sequences. The human antibodies of the present invention may include amino acid residues not encoded by human immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). As used herein the term “chimeric antibody" refers to an antibody which comprises a VH domain and a VL domain of a non-human antibody, and a CH domain and a CL domain of a human antibody. As used herein, the terms "monoclonal antibody", "monoclonal Ab", "monoclonal antibody composition", "mAb", or the like, as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. Monoclonal antibodies can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique, the human B-cell hybridoma technique and the EBV-hybridoma technique. As used herein, the term "Fab" denotes an antibody fragment having a molecular weight of about 50,000 and antigen binding activity, in which about a half of the N-terminal side of H chain and the entire L chain, among fragments obtained by treating IgG with a protease, papaine, are bound together through a disulfide bond. As used herein, the term "F(ab')2" refers to an antibody fragment having a molecular weight of about 100,000 and antigen binding activity, which is slightly larger than the Fab bound via a disulfide bond of the hinge region, among fragments obtained by treating IgG with a protease, pepsin. As used herein, the term "Fab' " refers to an antibody fragment having a molecular weight of about 50,000 and antigen binding activity, which is obtained by cutting a disulfide bond of the hinge region of the F(ab')2. As used herein, the term "single chain Fv" ("scFv") polypeptide is a covalently linked VH:VL heterodimer which is usually expressed from a gene fusion including VH and VL encoding genes linked by a peptide-encoding linker. As used herein, the term "dsFv" is a VH:VL heterodimer stabilised by a disulfide bond. Divalent and multivalent antibody fragments can form either spontaneously by association of monovalent scFvs, or can be generated by coupling monovalent scFvs by a peptide linker, such as divalent sc(Fv)2. As used herein, the term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. As used herein, the term “specificity” refers to the ability of an antibody to detectably bind an epitope presented on an antigen, such as Cath-D, while having relatively little detectable reactivity with non-Cath-D proteins or structures. Specificity can be relatively determined by binding or competitive binding assays, using, e.g., Biacore instruments, as described elsewhere herein. Specificity can be exhibited by, e.g., an about 10:1, about 20:1, about 50:1, about 100:1, 10.000:1 or greater ratio of affinity/avidity in binding to the specific antigen versus nonspecific binding to other irrelevant molecules (in this case the specific antigen is Cath-D). Many different competitive binding assay format(s) which can be used include, but are not limited to, competitive assay systems using techniques such western blots, radioimmunoassays, ELISA, “sandwich” immunoassays, immunoprecipitation assays, precipitin assays, gel diffusion precipitin assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and complement-fixation assays. Such assays are routine and well known in the art (see, e.g., Ausubel et al., eds, 1994 Current Protocols in Molecular Biology, Vol. 1, John Wiley & sons, Inc., New York). For example, the BIACORE® (GE Healthcare, Piscaataway, NJ) is one of a variety of surface plasmon resonance assay formats that are routinely used to epitope bin panels of monoclonal antibodies. Additionally, routine cross-blocking assays such as those described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane, 1988, can be performed. As used herein, the term “affinity”, as used herein, means the strength of the binding of an antibody to an epitope. The affinity of an antibody is given by the dissociation constant Kd, defined as [Ab] x [Ag] / [Ab-Ag], where [Ab-Ag] is the molar concentration of the antibody- antigen complex, [Ab] is the molar concentration of the unbound antibody and [Ag] is the molar concentration of the unbound antigen. The affinity constant Ka is defined by 1/Kd. Preferred methods for determining the affinity of mAbs can be found in Harlow, et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988), Coligan et al., eds., Current Protocols in Immunology, Greene Publishing Assoc. and Wiley Interscience, N.Y., (1992, 1993), and Muller, Meth. Enzymol. 92:589-601 (1983), which references are entirely incorporated herein by reference. One preferred and standard method well known in the art for determining the affinity of mAbs is the use of Biacore instruments. As used herein, the term "neutralizing antibody” refers to an antibody that blocks or reduces at least one activity of a polypeptide comprising the epitope to which the antibody specifically binds. A neutralizing antibody reduces Cathepsin D biological activity in in cellulo and/or in vivo tests. As used herein, the term “amino-acid sequence” has its general meaning and is a sequence of amino acids that confers to a protein its primary structure. According to the invention, the amino-acid sequence may be modified with one, two or three conservative amino acid substitutions, without appreciable loss of interactive binding capacity. By “conservative amino acid substitution”, it is meant that an amino acid can be replaced with another amino acid having a similar side chain. Families of amino acids having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., glycine, cysteine, alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta- branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). According to the invention a first amino-acid sequence having at least 70% of identity with a second amino-acid sequence means that the first sequence has 70; 71; 72; 73; 74; 75; 76; 77; 78; 79; 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; or 99% of identity with the second amino acid sequence. Amino-acid sequence identity is typically determined using a suitable sequence alignment algorithm and default parameters, such as BLAST P (Karlin and Altschul, 1990). According to the invention, the “identity” is calculated by comparing two aligned sequences in a comparison window. The sequence alignment allows determining the number of positions (nucleotides or amino acids) in common for the two sequences in the comparison window. The number of positions in common is therefore divided by the total number of positions in the comparison window and multiplied by 100 to obtain the identity percentage. The determination of the identity percentage of sequence can be made manually or thanks to well-known computer programs. As used herein, the terms “purified” and “isolated” relate to the antibody of the invention and mean that the antibody is present in the substantial absence of other biologic macromolecules of the same type. The term “purified” as used here means preferably that at least 75 % in weight, more preferably at least 85% in weight, even more preferably at least 95% in weight, and the more preferably at least 98% in weight of antibody, compared to the total weight of macromolecules present. As used herein, the term "nucleic acid molecule" has its general meaning in the art and refers to a DNA or RNA molecule. As used herein, the term “tumor-associated macrophages” also known as macrophages or Tumor-associated macrophages is a type of blood-borne phagocytes, derived from circulating monocytes or resident tissue macrophages. Their complex roles in carcinogenesis generally lead to disease progression in many cancers, which share some similar pathological mechanisms. There are two different subpopulations of activated macrophages within tumor microenvironment. The first type, known as classically activated macrophages (M1 macrophages or TAM-M1), are activated by lipopolysaccharides (LPS) or by double signals from interferon (IFN)-γ and tumor necrosis factor-α (TNF-α). This first type of macrophage is able to kill microorganisms and tumor cells. The second type of macrophages is known as immunosuppressive macrophages (M2 macrophages or TAM-2). Exposure to IL-4, IL-13, vitamin D3, glucocorticoids or transforming growth factor-β (TGF-β) decreases macrophage antigen-presenting capability and up-regulates the expression of macrophage mannose receptors (MMR, also known as CD206), scavenger receptors (SR-A, also known as CD204), dectin-1 and DC-SIGN.9 M2-polarized macrophages exhibit an IL-12low, IL-23low, IL- 10high phenotype. This second type of macrophage plays an important role in stroma formation, tissue repair, tumor growth, angiogenesis and immunosuppression. In breast cancer, TAMs are the most abundant inflammatory cells and are typically M2-polarized with suppressive capacity (1) that stems from their enzymatic activities and production of anti-inflammatory cytokines, such as TGFβ (Fuxe et al., Semin Cancer Biol, 2012, 22:455-461). High TAM levels have been associated with poorer BC outcomes (Zhao et al., Oncotarget, 2017, 8:30576-86. Therefore, several strategies are currently under investigation, such as the suppression of TAM recruitment, their depletion, or the switch from the pro-tumor M2 to the anti-tumor M1 phenotype in patients with TNBC (Georgoudaki et al., Cell Reports, 2016, 15:2000-11). As used herein, the term “natural killer cells” (NK cells or large granular lymphocytes) has its general meaning in the art and refers to a group of immune cells that belong to the innate lymphoid cells (ILC). NK cells can be identified by virtue of certain characteristics and biological properties, such as the expression of specific surface antigens including CD56 and/or NKp46 for human NK cells, the absence of the alpha/beta or gamma/delta TCR complex on the cell surface, the ability to bind to and kill cells that fail to express “self” MHC/HLA antigens by the activation of specific cytolytic machinery, the ability to kill tumor cells or other diseased cells that express a ligand for NK activating receptors, and the ability to release protein molecules called cytokines (such as cytolytic enzymes-granzyme B and perforin, and the anti- tumor cytokine TNFα) that stimulate the immune response. As used herein, the term "dendritic cell" or “DC” refers to a sub-type of antigen presenting cells that are characterized at the morphological level by numerous membrane processes that extend out from the main cell body (similar to dendrites on neurons) and at the biochemical level by cell surface expression of MHC class II molecules and lack of expression of one or more of CD3, CD14, CD19, CD56 and/or CD66b. Subsets of dendritic cells express on their cell surface CDlA, CDlC, CD50, CD54, CD58, CD102, CD80 and/or CD86. Some DCs also express toll-like receptors 2, 3, 4, 7 and/or 9. DCs encompass plasmacytoid dendritic cells (pDCs) and conventional dendritic cells (cDCs). pDCs are the main producer of type I (α/β) and III (λ) interferons (IFNs) upon the sensing of viral-type stimuli. cDCs are the most efficient cells for T cell priming and are further classified into cDC1s and cDC2s. As used herein, the term “T cells” has its general meaning in the art and represent an important component of the immune system that plays a central role in cell-mediated immunity. T cells are known as conventional lymphocytes as they recognize the antigen with their TCR (T cell receptor for the antigen) with presentation or restriction by molecules of the complex major histocompatibility. There are several subsets of T cells each having a distinct function such as CD8+ T cells, CD4+ T cells, Gamma delta T cells, and Tregs. As used herein, the term “CD8+ T cell” has its general meaning in the art and refers to a subset of T cells which express CD8 on their surface. They are MHC class I-restricted, and function as cytotoxic T cells. “CD8+ T cells” are also called cytotoxic T lymphocytes (CTL), T-killer cells, cytolytic T cells, or killer T cells. CD8 antigens are members of the immunoglobulin supergene family and are associative recognition elements in major histocompatibility complex class I-restricted interactions. As used herein, the term “tumor infiltrating CD8+ T cell” refers to the pool of CD8+ T cells of the patient that have left the blood stream and have migrated into a tumor. As used herein, the term “CD4+ T cells” (also called T helper cells or TH cells) refers to T cells which express the CD4 glycoprotein on their surfaces and which assist other white blood cells in immunologic processes, including maturation of B cells into plasma cells and memory B cells, and activation of cytotoxic T cells and macrophages. CD4+ T cells become activated when they are presented with peptide antigens by MHC class II molecules, which are expressed on the surface of antigen-presenting cells (APCs). Once activated, they divide rapidly and secrete cytokines that regulate or assist in the active immune response. These cells can differentiate into one of several subtypes, including TH1, TH2, TH3, TH17, TH9, TFH or Treg, which secrete different cytokines to facilitate different types of immune responses. Signaling from the APC directs T cells into particular subtypes. In addition to CD4, the TH cell surface biomarkers known in the art include CXCR3 (Th1), CCR4, Crth2 (Th2), CCR6 (Th17), CXCR5 (Tfh) and as well as subtype-specific expression of cytokines and transcription factors including T-bet, GATA3, EOMES, RORγT, BCL6 and FoxP3. As used herein, the term “exhausted T cells” or “exhaustion of T cells” has its general meaning in the art and refers to an altered differentiation state of T cells characterized by a loss of effector functions, sustained upregulation and co-expression of multiple inhibitory receptors, altered expression of inhibitory markers such as PD-1, PD-L1, LAG-3, CTLA4 and TIM-3 as well as impairment in their ability to release pro-inflammatory cytokines (IFNγ and TNFα). Exhaustion commonly occurs in the tumour microenvironment where T cells suffer a loss of their cytotoxic function and become ineffective in 12mmunoly to kill cancerous cells. As used herein, the term “Effector functions” refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation. As used herein, the term “antibody-dependent cell-mediated cytotoxicity” or “ADCC” has its general meaning in the art, and refers to a cell-mediated reaction in which non- specific cytotoxic cells that express Fc receptors (FcRs) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. Non-specific cytotoxic cells that mediate ADCC include natural killer (NK) cells, macrophages, monocytes, neutrophils, and eosinophils. As used herein, the term “Complement-dependent cytotoxicity” or “CDC” has its general meaning in the art, and refers to mechanisms when the protein C1q binds to antibody, which triggers the complement cascade, resulting in the formation of a membrane attack complex (MAC) on the surface of target cells, and further resulting in a classical pathway of complement activation. As used herein, the term “Antibody-dependent cellular phagocytosis “or “ADCP” has its general meaning in the art, and refers to process in which an antibody eliminates binding target and initiates phagocytosis by linking its Fc domain to a specific receptor on the phagocytic cell. Unlike ADCC, ADCP can mediate monocytes, macrophages, neutrophils and dendritic cells via FcγRIIa, FcγRI and FcγRIIIa As used herein, the terms “treating” or “treatment” refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subject at risk of contracting the disease or suspected to have contracted the disease as well as subject who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment. By “therapeutic regimen” is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a subject during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a “loading regimen”, which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase “maintenance regimen” or “maintenance period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a subject during treatment of an illness, e.g., to keep the subject in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., pain, disease manifestation, etc.]). As used herein, a “therapeutically effective amount” is intended for a minimal amount of active agent which is necessary to impart therapeutic benefit to a patient. For example, a “therapeutically effective amount of the active agent” to a patient is an amount of the active agent that induces, ameliorates or causes an improvement in the pathological symptoms, disease progression, or physical conditions associated with the disease affecting the patient. It will be understood that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts. For example, it is well known within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day. Preferably, the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient. An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 100 mg/kg of body weight per day. As used herein the terms “administering” or “administration” refer to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g., the nanobody or polypeptide according to the invention) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art. When a disease, or a symptom thereof, is being treated, administration of the substance typically occurs after the onset of the disease or symptoms thereof. When a disease or symptoms thereof, are being prevented, administration of the substance typically occurs before the onset of the disease or symptoms thereof. As used herein, the terms “combined treatment”, “combined therapy” or “therapy combination” refer to a treatment that uses more than one medication. The combined therapy may be dual therapy or bi-therapy. As used herein, the term “administration simultaneously” refers to administration of 2 active ingredients by the same route and at the same time or at substantially the same time. The term “administration separately” refers to an administration of 2 active ingredients at the same time or at substantially the same time by different routes. The term “administration sequentially” refers to an administration of 2 active ingredients at different times, the administration route being identical or different As used herein, the term “Pharmaceutically” or “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate. A pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. As used herein, the term “abnormal cell growth” and “hyperproliferative disorders or diseases” are used interchangeably in this application and refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). In the context of the invention the hyperproliferative diseases refers to diseases having an overexpression of cathepsin-D. Typically, hyperproliferative diseases are selected but not limited to, cancer (e.g. breast cancer, renal cancer etc), skin disorders (e.g. psoriasis, wound healing), inflammatory diseases (e.g. inflammatory bowel disease). As used herein, the term “cancer” or “tumor” refers to a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body. As used herein, the term “stroma “ or “microenvironment” has its general meaning in the art and refers to extracellular matrix and specialized connective tissue cells, including fibroblasts and mesenchymal stromal cells. Tumors have stroma and require stroma for nutritional support and the removal of waste products, but stromal content can vary markedly in different types of cancers. Antibodies of the invention: In the context of the invention, inventors have generated fully human anti-Cath-D single-chain variable antibody fragment (scFv). The inventors have cloned and characterized the variable domain of the light and heavy chains of said antibody, and thus determined the complementary determining regions (CDRs) domain of said antibody as described in Table 1: ScFv F1M1 Sequence (defined by IMGT unique numbering for IgG) Domains VH EVQLVESGGSLVKPGGSLRLSCAASGFTFSNNYMNWVRQAPGK GLEWISYISGSSRYISYADFVKGRFTISRDNAKNSLYLQMNSLRAE DTAVYYCVRSSNSGGMDVWGRGTLVTVSS (SEQ ID NO: 1) H-CDR1 GFTFSNNY (SEQ ID NO: 2) H-CDR2 ISGSSRYI (SEQ ID NO: 3) H-CDR3 VRSSNSGGMDV (SEQ ID NO: 4) VL QSVLTQPASVSGSPGQSITISCAGTSSDVGGYYGVSWYQQHPGKA PKLMIYYDSNRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYC SSYTSNSARVFGGGTKLAVL (SEQ ID NO: 5) L-CDR1 SSDVGGYYG (SEQ ID NO: 6) L-CDR2 YDS L-CDR3 SSYTSNSARV (SEQ ID NO: 7) Table 1: Sequences of scFv F1M1 antibody. In a first aspect, the invention relates to an isolated anti-Cathepsin-D antibody comprising (F1M1): (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises: a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7. In some embodiments, the anti-Cathepsin-D antibody according to the invention comprises H-CDR1 set forth as SEQ ID NO:2, H-CDR2 set forth as SEQ ID NO:3, H-CDR3 set forth as SEQ ID NO:4, L-CDR1 set forth as SEQ ID NO:6, L-CDR2 set forth as YDS, and L-CDR3 set forth as SEQ ID NO:7. In some embodiments, the anti-Cathepsin-D antibody according to the invention comprises: (a) a heavy chain wherein the variable domain has at least 70% of identity with a sequence set forth as SEQ ID NO:1 and/or (b) a light chain wherein the variable domain has at least 70% of identity with a sequence set forth as SEQ ID NO:5. In some embodiments, the anti-Cathepsin-D antibody according to the invention comprises: (a) a heavy chain wherein the variable domain has at least 70% of identity with a sequence set forth as SEQ ID NO:1 and comprises H-CDR1 set forth as SEQ ID NO:2, H-CDR2 set forth as SEQ ID NO:3, H-CDR3 set forth as SEQ ID NO:4, and/or (b) a light chain wherein the variable domain has at least 70% of identity with a sequence set forth as SEQ ID NO:5 and comprises L-CDR1 set forth as SEQ ID NO:6, L-CDR2 set forth as YDS, and L-CDR3 set forth as SEQ ID NO:7. In some embodiments, the anti-Cathepsin-D antibody according to the invention comprises (a) a heavy chain wherein the variable domain has a sequence set forth as SEQ ID NO:1 and/or (b) a light chain has a heavy chain wherein the variable domain has a sequence set forth as SEQ ID NO:5. In some embodiments, the anti-Cathepsin-D antibody according to the invention comprises (a) a heavy chain wherein the variable domain has a sequence set forth as SEQ ID NO:1 and (b) a light chain wherein the variable domain has a sequence set forth as SEQ ID NO:5 (“F1M1 antibody”). In some embodiments, the human anti-Cathepsin-D antibody according to the invention is a neutralizing antibody. In particular embodiment, the anti-Cathepsin-D antibody according to the invention is a chimeric anti-Cathepsin-D antibody, particularly a chimeric mouse/human antibody. In some embodiments, the human chimeric antibody of the present invention can be produced by obtaining nucleic sequences encoding VL and VH domains as previously described, constructing a human chimeric antibody expression vector by inserting them into an expression vector for animal cell having genes encoding human antibody CH and human antibody CL, and expressing the coding sequence by introducing the expression vector into an animal cell. As the CH domain of a human chimeric antibody, it may be any region which belongs to human immunoglobulin, but those of IgG class are suitable and any one of subclasses belonging to IgG class, such as IgG1, IgG2, IgG3 and IgG4, can also be used. Also, as the CL of a human chimeric antibody, it may be any region which belongs to Ig, and those of kappa class or lambda class can be used. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art (See Morrison SL. Et al. (1984) and patent documents US5,202,238; and US5,204, 244). In particular embodiment, the anti-Cathepsin-D antibody according to the invention is a human anti-Cathepsin-D antibody. Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, cur. Opin. Pharmacol. 5; 368-74 (2001) and lonberg, cur. Opin.Immunol.20; 450-459 (2008). Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal’s chromosomes. In such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated. For review of methods for obtaining human antibodies from transgenic animals, see Lonberg, Nat.Biotech.23;1117-1125 (2005). See also, e.g., U.S. Patent Nos. 6,075,181 and 6,150,584 describing XENOMOUSETM technology; U.S. Patent No. 5,770,429 describing HUMAB® technology; U.S. Patent No. 7,041,870 describing K-M MOUSE® technology, and U.S. Patent Application publication No. US 2007/0061900, describing VELOCIMOUSE® technology. Human variable regions from intact antibodies generated by such animals may be further modified, e.g., by combining with a different human constant region Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol., 13: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp.51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al., J. Immunol., 147: 86(1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al., Proc. Natl. Acad. Sci. USA, 103:3557-3562 (2006). Additional methods include those described, for example, in U.S. Patent No. 7,189,826 (describing production of monoclonal human igM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas). Human hybridoma technology (Trioma technology) is also described in Vollmers and Brandlein,, Histology and Histopathology, 20(3):927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3):185-91 (2005). Fully human antibodies can also be derived from phage- display libraries (as disclosed in Hoogenboom et al., 1991, J. Mol. Biol.227:381; and Marks et al., 1991, J. Mol. Biol. 222:581). Phage display techniques mimic immune selection through the display of antibody repertoires on the surface of filamentous bacteriophage, and subsequent selection of phage by their binding to an antigen of choice. One such technique is described in PCT publication No. WO 99/10494. Human antibodies described herein can also be prepared using SCID mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization. Such mice are described in, for example, U.S. Patent Nos.5,476,996 and 5,698,767 to Wilson et al. In a particular embodiment, the anti-Cathepsin-D antibody of the invention is an antibody fragment selected from the group consisting of Fab, F(ab’)2, Fab’, dsFv, diabodies and scFv. In a particular embodiment, the anti-Cathepsin-D antibody of the invention is scFv. The scFv of the present invention can be produced by obtaining cDNA encoding the VH and VL domains as previously described, constructing DNA encoding scFv, inserting the DNA into an expression vector for prokaryote, or an expression vector for eukaryote, and then introducing the expression vector into a prokaryote or eukaryote (as appropriate) to express the scFv. To generate a humanized scFv fragment, a well-known technology called CDR grafting may be used, which involves selecting the complementary determining regions (CDRs) from a donor scFv fragment, and grafting them onto a human scFv fragment framework of known three dimensional structure (see, e. g., W098/45322; WO 87/02671; US5,859,205; US5,585,089; US4,816,567; EP0173494). In some embodiments, the anti-Cathepsin-D antibody of the invention is a monoclonal antibody and more particularly a human monoclonal antibody. The antibody of the present invention may be of any isotype. IgGl and IgG3 are isotypes that mediate such effectors functions as ADCC or CDC, when IgG2 and IgG4 don’t or in a lower manner. Either of the human light chain constant regions, kappa or lambda, may be used. If desired, the class of a monoclonal antibody of the present invention may be switched by known methods. Typical, class switching techniques may be used to convert one IgG subclass to another, for instance from IgG1 to IgG2. Thus, the effector function of the monoclonal antibodies of the present invention may be changed by isotype switching to, e.g., an IgGl, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM antibody for various therapeutic uses. In some embodiments, the anti-Cathepsin-D antibody is a monoclonal IgG, and particularly a human monoclonal IgG antibody, and more particularly, a human monoclonal IgG1 antibody In some embodiments, the anti-Cathepsin-D antibody of the present invention is a full- length antibody and thus comprises an Fc region. In some embodiment, the anti-Cathepsin-D antibody of the present invention comprises an Fc region. In some embodiments, the isolated anti-Cathepsin-D antibody of the invention is engineered in order to improve its properties. Engineered antibodies of the invention include those in which modifications have been made to framework residues within VH and/or VL, e.g. to improve the properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody. For example, one approach is to “backmutate” one or more framework residues to the corresponding germline sequence. More specifically, an antibody that has undergone somatic mutation may contain framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is derived. To return the framework region sequences to their germline configuration, the somatic mutations can be ”backmutated” to the germline sequence by, for example, site-directed mutagenesis or PCR- mediated mutagenesis. Such “backmutated” antibodies are also intended to be encompassed by the inventio”. Another type of framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell - epitopes to thereby reduce the potential immunogenicity of the antibody. This approach is also referred to as "deimmunization" and is described in further detail in U.S. Patent Publication No. 20030153043 by Carr et al. In some embodiments, the glycosylation of the antibody of the invention is modified. Glycosylation can be altered to, for example, increase the affinity of the antibody for the antigen. Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site. Such aglycosylation may increase the affinity of the antibody for antigen. Such an approach is described in further detail in U.S. Patent Nos.5,714,350 and 6,350,861 by Co et al. In particular embodiment, the antibody of the invention is aglycosylated. In another embodiment, the antibody is modified to increase its biological half-life. Various approaches are possible. For example, one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Patent No. 6,277,375 by Ward. Alternatively, to increase the biological half-life, the antibody can be altered within the CH1 or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Patent Nos.5,869,046 and 6,121 ,022 by Presta et al. Antibodies with increased half live and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the foetus (Guyer et al., J. Immunol. 117:587 (1976) and Kim et al., J. immunol. 24:249 (1994)), are described in US2005/0014934A1 (Hinton et al.). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn. Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311,312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424, or 434, e.g., substitutions of Fc region residue 434 (US Patent No.7,371,826). Another modification of the antibodies herein that is contemplated by the invention is pegylation. An antibody can be pegylated to, for example, increase the biological (e.g., serum) half-life of the antibody. To pegylate an antibody, the antibody, or fragment thereof, typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment. The pegylation can be carried out by an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer). As used herein, the term "polyethylene glycol" is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (C1- C10) alkoxy- or aryloxy- polyethylene glycol or polyethylene glycol-maleimide. In certain embodiments, the antibody to be pegylated is an aglycosylated antibody. Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention. See for example, EP0154316 by Nishimura et al. and EP0401384 by Ishikawa et al. Another modification of the antibodies that is contemplated by the invention is a conjugate or a protein fusion of at least the antigen-binding region of the antibody of the invention to serum protein, such as human serum albumin or a fragment thereof to increase half-life of the resulting molecule. Such approach is for example described in Ballance et al. EP0322094. Another possibility is a fusion of at least the antigen-binding region of the antibody of the invention to proteins capable of binding to serum proteins, such human serum albumin to increase half-life of the resulting molecule. Such approach is for example described in Nygren et al., EP 0486525. Polysialytion is another technology, which uses the natural polymer polysialic acid (PSA) to prolong the active life and improve the stability of therapeutic peptides and proteins. PSA is a polymer of sialic acid (a sugar). When used for protein and therapeutic peptide drug delivery, polysialic acid provides a protective microenvironment on conjugation. This increases the active life of the therapeutic protein in the circulation and prevents it from being recognized by the immune system. The PSA polymer is naturally found in the human body. It was adopted by certain bacteria which evolved over millions of years to coat their walls with it. These naturally polysialylated bacteria were then able, by virtue of molecular mimicry, to foil the body's defense system. PSA, nature's ultimate stealth technology, can be easily produced from such bacteria in large quantities and with predetermined physical characteristics. Bacterial PSA is completely non-immunogenic, even when coupled to proteins, as it is chemically identical to PSA in the human body. In some embodiments, the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector functions of the antibody. For example, one or more amino acids can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen-binding ability of the parent antibody. The effector ligand to which affinity is altered can be, for example, an Fc receptor or the C1 component of complement. This approach is described in further detail in U.S. Patent Nos.5,624,821 and 5,648,260, both by Winter et al. In some embodiments, the hinge region of CH1 is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Patent No. 5,677,425 by Bodmer et al. The number of cysteine residues in the hinge region of CH1 is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody. In some embodiments, the Fc region is modified to increase the ability of the antibody to mediate antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fc receptor by modifying one or more amino acids. This approach is described further in PCT Publication WO 00/42072 by Presta. Moreover, the binding sites on human IgGI for FcγRI, FcγRII, FcγRIII and FcRn have been mapped and variants with improved binding have been described (see Shields, R. L. et al., 2001 J. Biol. Chen.276:6591- 6604, WO2010106180). The inventors have particularly designed an Fc-optimized F1M1 (“F1M1-Fc+”), in which the S239D, H268F, S324T, I332E mutations increase the binding affinity to CD16a (FcγRIIIA) to enhance NK cell activation, ADCC, ADCP and CDC. The inventors have particularly designed another Fc-variant F1M1 (“F1M1-Fc-”), in which the L234A, L235A and P329G mutations prevent binding to all Fcγ receptors including CD16a (FcγRIIIA) in order to prevent Fc-mediated cytotoxicity (ADCC, ADCP, ACDC). Thus, in some embodiments, the Fc region comprises at least 1, 2, 3 or 4 mutations selected from the group consisting of but not limited to: S239D, H268F, S324T and I332E. Thus, in some embodiments, the Fc region comprises at least 1, 2, 3 or 4 mutations selected from the group consisting of but not limited to: S239D, H268F, S324T and I332E. Thus in particular embodiment, the anti-Cathepsin-D antibody of the invention comprises a mutated Fc region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E. In other words, in some embodiments, the anti-Cathepsin-D antibody of the invention comprises a mutated Fc region comprising the following mutations S239D, H268F, S324T, I332E (“F1M1-Fc+”). Thus, in some embodiments, the anti-Cathepsin-D antibody of the invention comprises: (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7, and (c) a Fc region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E (“F1M1-Fc+”). In some embodiments, the anti-Cathepsin-D antibody of the invention comprise: (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5, and (c) a mutated Fc region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E (“F1M1-Fc+”). In particular embodiment, the anti-Cathepsin-D antibody of the invention comprises a mutated Fc region comprising at least 1, 2 or 3 mutations selected from the group consisting of L234A, L235A and P329G. In other words, in some embodiments, the anti-Cathepsin-D antibody of the invention comprises a mutated Fc region comprising the following mutations L234A, L235A and P329G (“F1M1-Fc-”). Thus, in some embodiments, the anti-Cathepsin-D antibody of the invention comprises : (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7, and (c) a Fc region comprising at least 1, 2 or 3 mutations selected from the group consisting of L234A, L235A and P329G (“F1M1-Fc-”). In some embodiments, the anti-Cathepsin-D antibody of the invention comprises: (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5, and (c) a mutated Fc region comprising at least 1, 2 or 3 mutations selected from the group consisting of L234A, L235A and P329G (“F1M1-Fc-”). It should be noted that the antibodies of the invention cross-react with rodents, and in particular murine (rat and mouse) Cathepsin-D or primate Cathepsin-D, which is of interest for preclinical evaluation and toxicological studies. It should be noted that no apparent toxicity was observed in syngenic mice or nude mice following “F1M1-Fc-“, “F1M1” and “F1M1-Fc+” treatment. It should be also noted that the antibodies of the invention specifically bind to Cathepsin- D, and do not bind with others aspartic proteases (e.g. cathepsin E, pepsinogen A and pepsinogen C). It should be further noted that the antibodies of the invention exhibit enhanced immunomodulatory activity. Indeed, it should be further noted that the antibodies of the invention inhibit the tumor recruitment of immunosuppressive tumor-associated macrophages M2, is able to enhance the activation of anti-tumor M1-polarized TAMs. In a particular embodiment, the anti-Cathepsin-D antibodies of the invention are able to promote the recruitment and maturation of conventional cDC1 dendritic cells. In a particular embodiment, the anti-Cathepsin-D antibodies of the invention are able to reduce the expression of exhaustion markers on CD4+ and CD8+ T cells, such as PD-L1 and LAG3, in tumors and draining lymph nodes. In a particular embodiment, the anti-Cathepsin-D antibodies of the invention are able to induce cytotoxicity, also known as the antibody-dependent cell-mediated cytotoxicity (ADCC), Complement-dependent cytotoxicity (CDC) and Antibody-dependent cellular phagocytosis (ADCP). ADCC is a mechanism of cell-mediated immune defense whereby an effector cell of the immune system actively lyses a target cell, whose membrane-surface antigens have been bound by specific antibodies. Typically, in the context of the invention, the anti-Cath-D antibodies as described above are able to activate NK cells (up-regulation of cytolytic enzymes-granzyme B and perforin, and the anti-tumor cytokine IFNγ suggesting the occurrence of ADCC in vivo. Typically, in the context of the invention, the Cathepsin-D antibodies F1M1 and F1M1-Fc+ as described above are able to activate NK cells (up-regulation of cytolytic enzymes-granzyme B and perforin, and the anti-tumor cytokine TNFα), suggesting the occurrence of ADCC in vivo. In a particular embodiment, the anti-Cathepsin-D antibodies of the invention are able to activate NK cells. In a particular embodiment, the human anti-Cathepsin-D antibody of the invention (F1M1-Fc+) is able to trigger the recruitment of NK cells in tumor. In some embodiments, the anti-Cathepsin-D antibody of the invention is able to activate NK cells and to induce cytotoxicity, wherein the antibody comprises: (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises: a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7. In some embodiments, the anti-Cathepsin-D antibody of the invention is able to activate NK cells and to induce cytotoxicity, wherein the antibody comprises: (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises: a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7, and (c) a FC region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E (“F1M1-Fc+”). In some embodiments, the anti-Cathepsin-D antibody of the invention is able to activate NK cells and to induce cytotoxicity, wherein the antibody comprises: (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5. In some embodiments, the anti-Cathepsin-D antibody of the invention is able to activate NK cells and to induce cytotoxicity, wherein the antibody comprises: (c) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (d) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5, and (e) A mutated Fc region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E (“F1M1-Fc+”). In some embodiments, the invention provides a multispecific antibody comprising a first antigen binding site from an anti-Cath-D antibody of the present invention described herein above and at least one second antigen binding site. In some embodiments, the second antigen-binding site is used for recruiting a killing mechanism such as, for example, by binding an antigen on a human effector cell as a BiTE (Bispecific T-Cell engager) antibody which is a bispecific scFv2 directed against target antigen and CD3 on T cells described in US7235641. As used herein, the term "effector cell" refers to an immune cell which is involved in the effector phase of an immune response, as opposed to the cognitive and activation phases of an immune response. Exemplary immune cells include a cell of a myeloid or lymphoid origin, for instance lymphocytes (such as B cells and T cells including cytolytic T cells (CTLs)), killer cells, natural killer cells, macrophages, monocytes, mast cells and granulocytes, such as neutrophils, eosinophils and basophils. Some effector cells express specific Fc receptors (FcRs) and carry out specific immune functions. In some embodiments, an effector cell is capable of inducing ADCC, such as a natural killer cell. For example, monocytes, macrophages, which express FcRs, are involved in specific killing of target cells and presenting antigens to other components of the immune system. In some embodiments, an effector cell may phagocytose a target antigen or target cell. The expression of a particular FcR on an effector cell may be regulated by humoral factors such as cytokines. An effector cell can phagocytose a target antigen or phagocytose or lyse a target cell. In some embodiments, the second antigen-binding site binds to an antigen on a human B cell, such as, e.g., CD19, CD20, CD21, CD22, CD23, CD46, CD80, CD138 and HLA-DR. Alternatively, in some embodiments, the second antigen-binding site binds to a different epitope of Cathepsin-D. In some embodiments, the second antigen-binding site binds to tumor-associated antigen. As used herein, the term “tumor-associated antigen” has its general meaning in the art and refers to an antigenic substance produced in tumor cells, i.e., it triggers an immune response in the host. Tumor-associated antigen (TAA) have elevated levels on tumors cells but are also expressed at lower levels on healthy cells. TAA include but are not limited to Human Epidermal Growth Factor Receptor-2 (HER2), Mucin-1 (MUC-1), carcinoembryonic antigen (CEA) and human telomerase reverse transcriptase (hTERT). In some embodiments, the second antigen-binding site binds to an angiogenic factor or other cancer-associated growth factor, such as a vascular endothelial growth factor (VEGF), a fibroblast growth factor (FGF), epidermal growth factor (EGF), angiogenin or a receptor of any of these, particularly receptors associated with cancer progression. In some embodiments, the second antigen-binding site binds to Fibroblast-activation protein (FAP). A further aspect of the invention refers to a cross-competing antibody which cross- competes for binding Cathepsin-D with the antibodies of the invention. In some embodiment, the cross-competing single-domain antibody of the present invention cross-competes for binding Cathepsin-D with the antibody comprising (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5 (“F1M1”) In some embodiment, the cross-competing single-domain antibody of the present invention cross-competes for binding Cathepsin-D with the antibody comprising (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5, and (c) a mutated Fc region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E (“F1M1-Fc+”). In some embodiment, the cross-competing single-domain antibody of the present invention cross-competes for binding Cathepsin-D with the antibody comprising (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5, and (c) a mutated Fc region comprising at least 1, 2, 3 mutations selected from the group consisting of L234A, L235A and P329G (“F1M1-Fc-”). As used herein, the term “cross-competes” refers to single-domain antibodies which share the ability to bind to a specific region of an antigen. In the present disclosure the single- domain antibody that “cross-competes" has the ability to interfere with the binding of another single-domain antibody for the antigen in a standard competitive binding assay. Such a single- domain antibody may, according to non-limiting theory, bind to the same or a related or nearby (e.g., a structurally similar or spatially proximal) epitope as the single-domain antibody with which it competes. Cross-competition is present if single-domain antibody A reduces binding of single-domain antibody B at least by 60%, specifically at least by 70% and more specifically at least by 80% and vice versa in comparison to the positive control which lacks one of said single-domain antibodies. As the skilled artisan appreciates competition may be assessed in different assay set-ups. One suitable assay involves the use of the Flow cytometry (using Flow cytometers and fluorescently-labeled cell suspension). Another possible assay for measuring cross-competition uses a cell-based ELISA approach. According to the present invention, the cross-competing antibody as above described retain the activity of the antibody of the invention (i.e able to inhibit the recruitment of immunosuppressive immune cells such as TAM, able to induce cytotoxicity (ADCC, CDC and ADCP) and to activate NK cells). In some embodiments, the cross-competing antibody as above described retain the activity of the antibody comprising: (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5 (“F1M1”). In some embodiments, the cross-competing antibody as above described retain the activity of the antibody comprising : (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5, and (c) a mutated Fc region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E (“F1M1- Fc+”). In some embodiments, the cross-competing antibody as above described retain the activity of the antibody comprising : (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5, and (c) an Fc region comprising at least 1, 2 or 3 mutations selected from the group consisting of L234A, L235A and P329G (“F1M1-Fc-”). Thus, in some embodiments, the cross-competing antibody of the present invention is anti-Cathepsin-D antibody, wherein said cross-competing antibody specifically binds to Cathepsin-D and is able to induce cytotoxicity and to activate NK cells. A further aspect of the invention refers to a chimeric antigen receptor (CAR) comprising an antigen binding domain of the anti-Cathepsin-D antibodies of the present invention. Typically, said chimeric antigen receptor comprises at least one VH and/or VL sequence of the antibody of the present invention. The chimeric antigen receptor of the present invention also comprises an extracellular hinge domain, a transmembrane domain, and an intracellular NK or T cell signaling domain. In particular embodiment, said chimeric antigen receptor comprises a VH sequence set forth as SEQ ID NO:1 and a VL sequence set forth as SEQ ID NO:5. As used herein, the term “chimeric antigen receptor” or “CAR” has its general meaning in the art and refers to a set of polypeptides, typically two in the simplest embodiments, which when in an immune effector cell, provides the cell with specificity for a target cell, typically a cancer cell, and with intracellular signal generation. In some embodiments, a CAR comprises at least an extracellular antigen binding domain, a transmembrane domain and a cytoplasmic signaling domain (also referred to herein as “an intracellular signaling domain”) comprising a functional signaling domain derived from a stimulatory molecule and/or costimulatory molecule as defined below. In some aspects, the set of polypeptides are contiguous with each other. In some embodiments, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signaling domain. In some embodiments, the stimulatory molecule is the zeta chain associated with the T cell receptor complex. In some embodiments, the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one costimulatory molecule as defined below. In some embodiments, the costimulatory molecule is chosen from the costimulatory molecules described herein, e.g., 4-1BB (i.e., CD137), CD27 and/or CD28. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a costimulatory molecule and a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising at least two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule.. In some embodiments, the CAR comprises an optional leader sequence at the amino-terminus (N-ter) of the CAR fusion protein. In some embodiments, the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen binding domain, wherein the leader sequence is optionally cleaved from the antigen binding domain (e.g., a scFv) during cellular processing and localization of the CAR to the cellular membrane. In particular aspects, CARs comprise fusions of single chain variable fragments (scFv) derived from monoclonal antibodies, fused to CD3-zeta a transmembrane domain and endodomain. In some embodiments, CARs comprise domains for additional co-stimulatory signaling, such as CD3-zeta, FcR, CD27, CD28, CD 137, DAP 10, and/or 0X40. In some embodiments, molecules can be co-expressed with the CAR, including co-stimulatory molecules, reporter genes for imaging (e.g., for positron emission tomography), gene products that conditionally ablate the T cells upon addition of a pro-drug, homing receptors, chemokines, chemokine receptors, cytokines, and cytokine receptors. A further aspect of the invention refers to a T-cell comprising the a chimeric antigen receptor of the invention (“CAR-T cell”). As used herein the term “CAR-T” has its general meaning in the art and refers to a T lymphocyte that has been genetically engineered to express a CAR. The definition of CAR T- cells encompasses all classes and subclasses of T-lymphocytes including CD4+ , CD8+ T cells, gamma delta T cells as well as effector T cells, memory T cells, regulatory T cells, and the like. The T lymphocytes that are genetically modified may be "derived" or "obtained" from the patient who will receive the treatment using the genetically modified T cells or they may be "derived" or "obtained" from a different patient. A further aspect of the invention refers to a NK-cell comprising the a chimeric antigen receptor of the invention (“CAR-NK cell”). As used herein the term “CAR-NK” refers to natural killer (NK) cells that has been genetically engineered to express a CAR. NK cells are defined as CD56+ and CD3– cells and are subdivided into cytotoxic and immunoregulatory. They are of great clinical interest because they contribute to the graft-vs-leukemia/graft-vs-tumor effect but are not responsible for graft- vs-host disease. NK cells can be generated from various sources such as umbilical cord blood, bone marrow, human embryonic stem cells, and induced pluripotent stem cells. However, tumors can escape the cytotoxicity of NK cells when they are directed against NKG2D ligands MICA and MICB (major histocompatibility complex class I chain-related protein A/B). Henceforth, preclinical research has been reported for CAR-modified primary human NK cells redirected against CD19, CD20, CD244, and HER2, as well as CAR-expressing NK-92 cells targeted to a wider range of cancer antigens. Primary NK cells engineered to express CARs have potential benefits compared to CAR-T cells. NK cells have spontaneous cytotoxic activity and can generate target cell death independent of tumor antigen, while T lymphocytes only kill their targets by a CAR-specific mechanism. Therefore, in the setting of antigen downregulation by tumor cells attempting to escape immune detection, NK cells would still be effective against tumor cells. In addition, primary human NK cells produce cytokines, such as interferon gamma, interleukin 3, and granulocyte-macrophage colony-stimulating factor, that differ from the proinflammatory cytokines produced by T cells that are responsible for the onset of cytokine release syndrome. Individual NK cells can survive after contacting and killing multiple target cells, possibly reducing the number of cells that need to be adoptively transferred (ie, the ex vivo stimulation and expansion of autologous or allogeneic lymphocytes, followed by reinfusion of the expanded lymphocyte population into the patient, in contrast to T cells). Furthermore, whereas the long-term persistence of CAR-T cells may maintain on-target, off- tumor toxicity such as the B cell aplasia seen with anti-CD19 CAR-T cells, mature NK cells are short lived and are expected to disappear after facilitating their anticancer effects. A further aspect of the invention refers to a tumor-associated macrophage (TAM) comprising the chimeric antigen receptor of the invention (“CAR-M cell”). As used herein the term “CAR-M” refers to tumor-associated macrophage that has been genetically engineered to express a CAR. Tumor-associated macrophages (TAMs) are the most abundant innate immune cells and constitute up to 50% of the cell mass within the tumor microenvironment (TME) of most solid tumors. Based on their ability to penetrate solid tumors and traffic through the TME, TAMs engineered with CAR constructs demonstrate sufficient potency. Similar to CAR-T, the core components of CAR-M contain an extracellular domain that provides specific recognition by a single-chain variable fragment (scFv) (eg, CD19 and HER2), a hinge domain, a transmembrane domain (mostly CD8), and an intracellular domain that presents dedicated downstream signalling (eg, CD3ζ, FcγR). Immunoconjugates of the invention: In some embodiments, the anti-Cathepsin-D antibody of the present invention is conjugated to a therapeutic moiety, i.e. a drug. The therapeutic moiety can be, e.g., a cytotoxin, a chemotherapeutic agent, a cytokine, an immunosuppressant, an immune stimulator, a lytic peptide, or a radioisotope. Such conjugates are referred to herein as an "antibody-drug conjugates" or "ADCs". Thus, in another aspect, the invention refers to an anti-Cathepsin-D antibody comprising : (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7, and wherein said anti-Cathepsin-D antibody is conjugated to a therapeutic moiety. In some embodiments, the anti-Cathepsin-D antibody of the invention comprises an Fc region comprising at least 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E. In some embodiments, the anti-Cathepsin-D antibody of the invention comprises an Fc region comprising at least 3 mutations selected from the group consisting of L234A, L235A and P329G. Thus, in another aspect, the invention refers to an anti-Cathepsin-D antibody comprising: (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7, and (c) a Fc region comprising at least 1, 2 or 3 mutations selected from the group consisting of L234A, L235A and P329G, and wherein said anti-Cathepsin-D antibody is conjugated to a therapeutic moiety In some embodiments, the therapeutic moiety is a cytotoxic moiety. In particular embodiments, the anti-Cathepsin-D antibody of the invention is conjugated to a cytotoxic moity, such that the resulting antibody-drug conjugate exerts a cytotoxic or cytostatic effect on a Cath-D-expressing cell (e.g., a Cath-D-expressing cancer cell) when taken up or internalized by the cell. Particularly suitable moieties for conjugation to antibodies are chemotherapeutic agents, prodrug converting enzymes, radioactive isotopes or compounds, or toxins. For example, an anti-Cath-D antibody can be conjugated to a cytotoxic moiety such as a chemotherapeutic agent or a toxin (e.g., a cytostatic or cytocidal agent such as, for example, abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin). Useful classes of cytotoxic moiety include, for example, antitubulin agents, auristatins, DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cis-platin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and-carboplatin), anthracyclines, antibiotics, antifolates, antimetabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, pre-forming compounds, purine antimetabolites, puromycins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like. Individual cytotoxic include, for example, an androgen, anthramycin (AMC), asparaginase, 5-azacytidine, azathioprine, bleomycin, busulfan, buthionine sulfoximine, camptothecin, carboplatin, carmustine (BSNU), CC-1065 (Li et al., Cancer Res.42:999-1004, 1982), chlorambucil, cisplatin, colchicine, cyclophosphamide, cytarabine, cytidine arabinoside, cytochalasin B, dacarbazine, dactinomycin (formerly actinomycin), daunorubicin, 34ibavirin34n, docetaxel, doxorubicin, an estrogen, 5-fluordeoxyuridine, etopside phosphate (VP-16), 5-fluorouracil, gramicidin D, hydroxyurea, idarubicin, ifosfamide, irinotecan, lomustine (CCNU), mechlorethamine, melphalan, 6-mercaptopurine, methotrexate, mithramycin, mitomycin C, mitoxantrone, nitroimidazole, paclitaxel, plicamycin, 35ibavirin35ne, streptozotocin, tenoposide (VM-26), 6-thioguanine, thioTEPA, topotecan, exatecan, deruxtecan, vinblastine, vincristine, and vinorelbine. Particularly suitable cytotoxic moiety include, for example, dolastatins (e.g., auristatin E, AFP, MMAF, MMAE), DNA minor groove binders (e.g., enediynes and lexitropsins), duocarmycins, taxanes (e.g., paclitaxel and docetaxel), puromycins, vinca alkaloids, CC-1065, SN-38 (7-ethyl-10-hydroxy-camptothein),, morpholino-doxorubicin, rhizoxin, cyanomorpholino-doxorubicin, echinomycin, combretastatin, netropsin, epothilone A and B, estramustine, cryptophysins, cemadotin, maytansinoids, discodermolide, eleutherobin, and mitoxantrone. In certain embodiments, a cytotoxic agent is a conventional chemotherapeutic such as, for example, doxorubicin, paclitaxel, melphalan, vinca alkaloids, methotrexate, mitomycin C or etoposide. In addition, potent agents such as CC-1065 analogues, calicheamicin, maytansine, analogues of dolastatin 10, rhizoxin, and palytoxin can be linked to an anti-Cath-D antibody. In specific variations, the cytotoxic or cytostatic moiety is auristatin E (also known in the art as dolastatin-10) or a derivative thereof. Typically, the auristatin E derivative is, e.g., an ester formed between auristatin E and a keto acid. For example, auristatin E can be reacted with paraacetyl benzoic acid or benzoylvaleric acid to produce AEB and AEVB, respectively. Other typical auristatin derivatives include AFP (dimethylvaline-valine-dolaisoleuine-dolaproine- phenylalanine-p-phenylenediamine), MMAF (dovaline-valine-dolaisoleunine-dolaproine- phenylalanine), and MAE (monomethyl auristatin E). The synthesis and structure of auristatin E and its derivatives are described in U.S. Patent Application Publication No. 20030083263; International Patent Publication Nos. WO 2002/088172 and WO 2004/010957; and U.S. Patent Nos.6,884,869; 6,323,315; 6,239,104; 6,034,065; 5,780,588; 5,665,860; 5,663,149; 5,635,483; 5,599,902; 5,554,725; 5,530,097; 5,521,284; 5,504,191; 5,410,024; 5,138,036; 5,076,973; 4,986,988; 4,978,744; 4,879,278; 4,816,444; and 4,486,414. In other variations, the cytotoxic moiety is a DNA minor groove binding agent. (See, e.g., U.S. Patent No. 6,130,237.) For example, in certain embodiments, the minor groove binding agent is a CBI compound. In other embodiments, the minor groove binding agent is an enediyne (e.g., calicheamicin). In certain embodiments, the cytotoxic moiety is an anti-tubulin agent. Examples of anti- tubulin agents include, for example, taxanes (e.g., Taxol® (paclitaxel), Taxotere® (docetaxel)), T67 (Tularik), vinca alkyloids (e.g., vincristine, vinblastine, vindesine, and vinorelbine), and dolastatins (e.g., auristatin E, AFP, MMAF, MMAE, AEB, AEVB). Other antitubulin agents include, for example, baccatin derivatives, taxane analogs (e.g., epothilone A and B), nocodazole, colchicine and colcimid, estramustine, cryptophysins, cemadotin, maytansinoids, combretastatins, discodermolide, and eleutherobin. In some embodiments, the cytotoxic agent is a maytansinoid, another group of anti-tubulin agents. For example, in specific embodiments, the maytansinoid is maytansine or DM-1 (ImmunoGen, Inc.; see also Chari et al., Cancer Res. 52:127-131, 1992). In other embodiments, the cytotoxic agent is an antimetabolite. The antimetabolite can be, for example, a purine antagonist (e.g., ibavirinne or mycophenolate mofetil), a dihydrofolate reductase inhibitor (e.g., methotrexate), acyclovir, gangcyclovir, zidovudine, vidarabine, ibavirine, azidothymidine, cytidine arabinoside, amantadine, dideoxyuridine, iododeoxyuridine, poscarnet, or trifluridine. In other embodiments, the cytotoxic agent is selected from the group consisting of topoisomerase inhibitors; ethidium bromide; emetine; dihydroxy anthracin dione; a tubulin- inhibitor such as maytansine or an analog or derivative thereof; an antimitotic agent such as monomethyl auristatin E or F or an analog or derivative thereof; dolastatin 10 or 15 or an analogue thereof; irinotecan or an analogue thereof; mitoxantrone; mithramycin; dactinomycin; 1-dehydrotestosterone; a glucocorticoid; procaine; tetracaine; lidocaine; propranolol; puromycin; calicheamicin or an analog or derivative thereof; an antimetabolite such as methotrexate, 6 mercaptopurine, 6 thioguanine, cytarabine, fludarabin, 5 fluorouracil, decarbazine, hydroxyurea, asparaginase, gemcitabine, or cladribine; an alkylating agent such as mechlorethamine, thioepa, chlorambucil, melphalan, carmustine (BSNU), lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, dacarbazine (DTIC), procarbazine, mitomycin C; a platinum derivative such as cisplatin or carboplatin; duocarmycin A, duocarmycin SA, rachelmycin (CC-1065), or an analog or derivative thereof; an antibiotic such as dactinomycin, bleomycin, daunorubicin, doxorubicin, idarubicin, mithramycin, mitomycin, mitoxantrone, plicamycin, anthramycin (AMC)); pyrrolo[2,l-c][l,4]- benzodiazepines (PDB); diphtheria toxin and related molecules such as diphtheria A chain and active fragments thereof and hybrid molecules, ricin toxin such as ricin A or a deglycosylated ricin A chain toxin, cholera toxin, a Shiga-like toxin such as SLT I, SLT II, SLT IIV, LT toxin, C3 toxin, Shiga toxin, pertussis toxin, tetanus toxin, soybean Bowman-Birk protease inhibitor, Pseudomonas exotoxin, alorin, saporin, modeccin, gelanin, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolacca americana proteins such as PAPI, PAPII, and PAP-S, momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, and enomycin toxins; ribonuclease (RNase); DNase I, Staphylococcal enterotoxin A; pokeweed antiviral protein; diphtherin toxin; and Pseudomonas endotoxin. In some embodiments, the antibody is conjugated to a nucleic acid or nucleic acid- associated molecule. In one such embodiment, the conjugated nucleic acid is a cytotoxic ribonuclease (RNase) or deoxy-ribonuclease (e.g., DNase I), an antisense nucleic acid, an inhibitory RNA molecule (e.g., a siRNA molecule) or an immunostimulatory nucleic acid (e.g., an immunostimulatory CpG motif-containing DNA molecule). In some embodiments, the antibody is conjugated to an aptamer or a ribozyme. In some embodiments, the antibody is conjugated to a radioisotope or to a radioisotope- containing chelate. For example, the antibody can be conjugated to a chelator linker, e.g. DOTA, DTPA or tiuxetan, which allows for the antibody to be complexed with a radioisotope. The antibody may also or alternatively comprise or be conjugated to one or more radiolabeled amino acids or other radiolabeled moleculesNon-limiting examples of radioisotopes include3H,14C,15N,35S,90Y,99Tc,125I,131I,186Re,213Bi,225Ac and227Th. For therapeutic purposes, a radioisotope emitting beta- or alpha-particle radiation can be used, e.g., 1311, 90Y, 211At, 212Bi, 67Cu, 186Re, 188Re, and 212Pb. In other embodiments, the anti-Cath-D antibody of the invention is conjugated to a pro- drug converting enzyme. The pro-drug converting enzyme can be recombinantly fused to the antibody or chemically conjugated thereto using known methods. Exemplary pro-drug converting enzymes are carboxypeptidase G2, β-glucuronidase, penicillin-V-amidase, penicillin-G-amidase, β-lactamase, β-glucosidase, nitroreductase and carboxypeptidase A. Techniques for conjugating therapeutic agents to proteins, and in particular to antibodies, are well-known. (See, e.g., Arnon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy,” in Monoclonal Antibodies And Cancer Therapy (Reisfeld et al. eds., Alan R. Liss, Inc., 1985); Hellstrom et al., “Antibodies For Drug Delivery,” in Controlled Drug Delivery (Robinson et al. eds., Marcel Deiker, Inc., 2nd ed.1987); Thorpe, “Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review,” in Monoclonal Antibodies ‘84: Biological And Clinical Applications (Pinchera et al. eds., 1985); “Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody In Cancer Therapy,” in Monoclonal Antibodies For Cancer Detection And Therapy (Baldwin et al. eds., Academic Press, 1985); and Thorpe et al., 1982, Immunol. Rev.62:119-58. See also, e.g., PCT publication WO 89/12624.) In other embodiments, the anti-Cath-D antibody of the invention is conjugated to a proteolyse targeting chimeric molecules or a molecular glue degraders. As used herein, the term “proteolyse targeting chimeric molecules”, also known as “PROTAC”, has its general meaning in the art and refers to a heterobifunctional molecule composed of two active domains and a linker, capable of removing specific unwanted proteins. PROTAC works by bringing together the E3 ligase with the target protein thus allowing its ubiquitination and degradation by the proteasome. As used herein, the term “molecular glue degraders” has its general meaning in the art and refers to monovalent compounds that orchestrate interactions between a target protein and an E3 ubiquitin ligase, prompting the proteasomal degradation of the former. Techniques for conjugating therapeutic agents such as PROTAC and molecular glue degraders, and in particular to antibodies, are well-known. (See, e.g., Conilh L, Sadilkova L, Viricel W, Dumontet C. Payload diversification: a key step in the development of antibody-drug conjugates. J Hematol Oncol.2023 Jan 17;16(1):3.; Dragovich PS. Degrader-antibody conjugates. Chem Soc Rev.2022 May 23;51(10):3886-3897.). Typically, the antibody-drug conjugate compounds comprise a linker unit between the drug unit and the antibody unit. In some embodiments, the linker is cleavable under intracellular conditions, such that cleavage of the linker releases the drug unit from the antibody in the intracellular environment. In yet other embodiments, the linker unit is not cleavable and the drug is released, for example, by antibody degradation. In some embodiments, the linker is cleavable by a cleaving agent that is present in the intracellular environment (e.g., within a lysosome or endosome or caveolea). The linker can be, e.g., a peptidyl linker that is cleaved by an intracellular peptidase or protease enzyme, including, but not limited to, a lysosomal or endosomal protease. In some embodiments, the peptidyl linker is at least two amino acids long or at least three amino acids long. Cleaving agents can include cathepsins B and D and plasmin, all of which are known to hydrolyze dipeptide drug derivatives resulting in the release of active drug inside target cells (see, e.g., Dubowchik and Walker, 1999, Pharm. Therapeutics 83:67-123). Most typical are peptidyl linkers that are cleavable by enzymes that are present in 191P4D12-expressing cells. Examples of such linkers are described, e.g., in U.S. Pat. No. 6,214,345, incorporated herein by reference in its entirety and for all purposes. In a specific embodiment, the peptidyl linker cleavable by an intracellular protease is a Val-Cit linker or a Phe-Lys linker (see, e.g., U.S. Pat. No.6,214,345, which describes the synthesis of doxorubicin with the Val-Cit linker). One advantage of using intracellular proteolytic release of the therapeutic agent is that the agent is typically attenuated when conjugated and the serum stabilities of the conjugates are typically high. In other embodiments, the cleavable linker is pH-sensitive, i.e., sensitive to hydrolysis at certain pH values. Typically, the pH-sensitive linker hydrolyzable under acidic conditions. For example, an acid-labile linker that is hydrolyzable in the lysosome (e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide, orthoester, acetal, ketal, or the like) can be used. (See, e.g., U.S. Pat. Nos. 5,122,368; 5,824,805; 5,622,929; Dubowchik and Walker, 1999, Pharm. Therapeutics 83:67-123; Neville et al., 1989, Biol. Chem.264:14653-14661.) Such linkers are relatively stable under neutral pH conditions, such as those in the blood, but are unstable at below pH 5.5 or 5.0, the approximate pH of the lysosome. In certain embodiments, the hydrolyzable linker is a thioether linker (such as, e.g., a thioether attached to the therapeutic agent via an acylhydrazone bond (see, e.g., U.S. Pat. No.5,622,929). In yet other embodiments, the linker is cleavable under reducing conditions (e.g., a disulfide linker). A variety of disulfide linkers are known in the art, including, for example, those that can be formed using SATA (N-succinimidyl-S-acetylthioacetate), SPDP (N- succinimidyl-3-(2-pyridyldithio)propionate), SPDB (N-succinimidyl-3-(2- pyridyldithio)butyrate) and SMPT (N-succinimidyl-oxycarbonyl-alpha-methyl-alpha-(2- pyridyl-dithio)toluene), SPDB and SMPT. (See, e.g., Thorpe et al., 1987, Cancer Res.47:5924- 5931; Wawrzynczak et al., In Immunoconjugates: Antibody Conjugates in Radioimagery and Therapy of Cancer (C. W. Vogel ed., Oxford U. Press, 1987. See also U.S. Pat. No.4,880,935.) In yet other specific embodiments, the linker is a malonate linker (Johnson et al., 1995, Anticancer Res.15:1387-93), a maleimidobenzoyl linker (Lau et al., 1995, Bioorg-Med-Chem. 3(10):1299-1304), or a 3′-N-amide analog (Lau et al., 1995, Bioorg-Med-Chem. 3(10):1305- 12). In yet other embodiments, the linker unit is not cleavable and the drug is released by antibody degradation. Typically, the linker is not substantially sensitive to the extracellular environment. As used herein, “not substantially sensitive to the extracellular environment,” in the context of a linker, means that no more than about 20 %, typically no more than about 15 %, more typically no more than about 10 %, and even more typically no more than about 5 %, no more than about 3 %, or no more than about 1 % of the linkers, in a sample of antibody-drug conjugate compound, are cleaved when the antibody-drug conjugate compound is present in an extracellular environment (e.g., in plasma). Whether a linker is not substantially sensitive to the extracellular environment can be determined, for example, by incubating with plasma the antibody-drug conjugate compound for a predetermined time period (e.g., 2, 4, 8, 16, or 24 hours) and then quantitating the amount of free drug present in the plasma. Techniques for conjugating molecules to antibodies, are well-known in the art (See, e.g., Arnon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy,” in Monoclonal Antibodies And Cancer Therapy (Reisfeld et al. eds., Alan R. Liss, Inc., 1985); Hellstrom et al., “Antibodies For Drug Delivery,” in Controlled Drug Delivery (Robinson et al. eds., Marcel Deiker, Inc., 2nd ed. 1987); Thorpe, “Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review,” in Monoclonal Antibodies '84: Biological And Clinical Applications (Pinchera et al. eds., 1985); “Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody In Cancer Therapy,” in Monoclonal Antibodies For Cancer Detection And Therapy (Baldwin et al. eds., Academic Press, 1985); and Thorpe et al., 1982, Immunol. Rev.62:119-58. See also, e.g., PCT publication WO 89/12624.) Typically, the nucleic acid molecule is covalently attached to lysines or cysteines on the antibody, through N- hydroxysuccinimide ester or maleimide functionality respectively. Methods of conjugation using engineered cysteines or incorporation of unnatural amino acids have been reported to improve the homogeneity of the conjugate (Axup, J.Y., Bajjuri, K.M., Ritland, M., Hutchins, B.M., Kim, C.H., Kazane, S.A., Halder, R., Forsyth, J.S., Santidrian, A.F., Stafin, K., et al. (2012). Synthesis of site-specific antibody-drug conjugates using unnatural amino acids. Proc. Natl. Acad. Sci. USA 109, 16101–16106.; Junutula, J.R., Flagella, K.M., Graham, R.A., Parsons, K.L., Ha, E., Raab, H., Bhakta, S., Nguyen, T., Dugger, D.L., Li, G., et al. (2010). Engineered thio-trastuzumab-DM1 conjugate with an improved therapeutic index to target humanepidermal growth factor receptor 2-positive breast cancer. Clin. Cancer Res.16, 4769– 4778.). Junutula et al. (2008) developed cysteine-based site-specific conjugation called ‘‘THIOMABs’’ (TDCs) that are claimed to display an improved therapeutic index as compared to conventional conjugation methods. Conjugation to unnatural amino acids that have been incorporated into the antibody is also being explored for ADCs; however, the generality of this approach is yet to be established (Axup et al., 2012). In particular the one skilled in the art can also envisage Fc-containing polypeptide engineered with an acyl donor glutamine-containing tag (e.g., Gin-containing peptide tags or Q- tags) or an endogenous glutamine that are made reactive by polypeptide engineering (e.g., via amino acid deletion, insertion, substitution, or mutation on the polypeptide). Then a transglutaminase, can covalently crosslink with an amine donor agent (e.g., a small molecule comprising or attached to a reactive amine) to form a stable and homogenous population of an engineered Fc-containing polypeptide conjugate with the amine donor agent being site- specifically conjugated to the Fc-containing polypeptide through the acyl donor glutamine- containing tag or the accessible/exposed/reactive endogenous glutamine (WO 2012059882). Example of techniques for conjugating molecules to antibodies, via some of the eight cysteines forming interchain disulfide bridges are described in the patent WO2015004400, incorporated herein by reference In particular embodiment, the anti-Cathepsin-D antibody of the invention is conjugated to at least one therapeutic moiety. In particular embodiment, the anti-Cathepsin-D antibody of the invention is conjugated to 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 therapeutic moiety. In other words, In some embodiments, the drug-to-antibody ratio (DAR) of the antibody-drug conjugate comprising the anti-Cathepsin-D antibody of the invention is between 1 to 10, and more particularly between 4 to 8. In other words, In some embodiments, the drug-to-antibody ratio (DAR) of the antibody-drug conjugate comprising the anti-Cathepsin-D antibody of the invention is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10, and more particularly is 8. As used herein, the term “Drug to Antibody Ratio” or “DAR” refers to the average number of drugs linked to each antibody. DAR is a key property used to measures the quality of ADC because it can significantly affect ADC efficacy. Nucleic acids, vectors, recombinant host cells of the invention A further object of the invention relates to a nucleic acid molecule encoding an anti- Cathepsin-D antibody according to the invention. More particularly the nucleic acid molecule encodes a heavy chain and/or a light chain of an anti-Cathepsin-D antibody of the present invention. Typically, said nucleic acid is a DNA or RNA molecule, which may be included in any suitable vector, such as a plasmid, cosmid, episome, artificial chromosome, phage or a viral vector. As used herein, the terms “vector”, “cloning vector” and “expression vector” mean the vehicle by which a DNA or RNA sequence (e.g. a foreign gene) can be introduced into a host cell, so as to transform the host and promote expression (e.g. transcription and translation) of the Introduced sequence. The terms “expression vector”, “expression construct” or “expression cassett” are used interchangeably throughout this specification and are meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed. So, a further aspect of the invention relates to a vector comprising a nucleic acid of the invention. Such vectors may comprise regulatory elements, such as a promoter, enhancer, terminator and the like, to cause or direct expression of said antibody upon administration to a subject. Examples of promoters and enhancers used in the expression vector for animal cell include early promoter and enhancer of SV40 (Mizukami T. et al. 1987), LTR promoter and enhancer of Moloney mouse leukemia virus (Kuwana Y et al.1987), promoter (Mason JO et al.1985) and enhancer (Gillies SD et al.1983) of immunoglobulin H chain and the like. Any expression vector for animal cell can be used, so long as a gene encoding the human antibody C region can be inserted and expressed. Examples of suitable vectors include pAGE107 (Miyaji H et al. 1990), pAGE103 (Mizukami T et al. 1987), pHSG274 (Brady G et al. 1984), pKCR (’'Hare K et al.1981), pSG1 beta d2-4-(Miyaji H et al.1990) and the like. Other examples of plasmids include replicating plasmids comprising an origin of replication, or integrative plasmids, such as for instance pUC, pcDNA, pBR, and the like. Other examples of viral vector include adenoviral, retroviral, herpes virus and AAV vectors. Such recombinant viruses may be produced by techniques known in the art, such as by transfecting packaging cells or by transient transfection with helper plasmids or viruses. Typical examples of virus packaging cells include PA317 cells, PsiCRIP cells, gPenv+ cells, 293 cells, etc. Detailed protocols for producing such replication-defective recombinant viruses may be found for instance in WO 95/14785, WO 96/22378, US 5,882,877, US 6,013,516, US 4,861,719, US 5,278,056 and WO 94/19478. The choice of a suitable expression vector for expression of the peptides or polypeptides of the invention will of course depend upon the specific host cell to be used, and is within the skill of the ordinary artisan. Expression requires that appropriate signals be provided in the vectors, such as enhancers/promoters from both viral and mammalian sources that may be used to drive expression of the nucleic acids of interest in host cells. Usually, the nucleic acid being expressed is under transcriptional control of a promoter. A “promote” refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene. Nucleotide sequences are operably linked when the regulatory sequence functionally relates to the DNA encoding the protein of interest (e.g., a single domain antibody). Thus, a promoter nucleotide sequence is operably linked to a given DNA sequence if the promoter nucleotide sequence directs the transcription of the sequence. A further aspect of the invention relates to a host cell which has been transfected, infected or transformed by a nucleic acid and/or a vector according to the invention. The term “transformation” means the introduction of a “foreign” (i.e. extrinsic or extracellular) gene, DNA or RNA sequence to a host cell, so that the host cell will express the introduced gene or sequence to produce a desired substance, typically a protein or enzyme coded by the introduced gene or sequence. A host cell that receives and expresses introduced DNA or RNA bas been “transformed”. The nucleic acids of the invention may be used to produce an antibody of the present invention in a suitable expression system. The term “expression system” means a host cell and compatible vector under suitable conditions, e.g. for the expression of a protein coded for by foreign DNA carried by the vector and introduced to the host cell. Common expression systems include E. coli host cells and plasmid vectors, insect host cells and Baculovirus vectors, and mammalian host cells and vectors. Other examples of host cells include, without limitation, prokaryotic cells (such as bacteria) and eukaryotic cells (such as yeast cells, mammalian cells, insect cells, plant cells, etc.). Specific examples include E.coli, Kluyveromyces or Saccharomyces yeasts, mammalian cell lines (e.g., Vero cells, CHO cells, 3T3 cells, COS cells, etc.) as well as primary or established mammalian cell cultures (e.g., produced from lymphoblasts, fibroblasts, non-human embryonic cells, epithelial cells, nervous cells, adipocytes, etc.). Examples also include mouse SP2/0-Ag14 cell (ATCC CRL1581), mouse P3X63-Ag8.653 cell (ATCC CRL1580), CHO cell in which a dihydrofolate reductase gene (hereinafter referred to as “DHFR gene”) is defective (Urlaub G et al; 1980), rat YB2/3HL.P2.G11.16Ag.20 cell (ATCC CRL1662, hereinafter referred to as “YB2/0 cell”), and the like. The present invention also relates to a method of producing a recombinant host cell expressing an antibody according to the invention, said method comprising the steps of: (i) introducing in vitro or ex vivo a recombinant nucleic acid or a vector as described above into a competent host cell, (ii) culturing in vitro or ex vivo the recombinant host cell obtained and (iii), optionally, selecting the cells which express and/or secrete said antibody. Such recombinant host cells can be used for the production of antibodies of the present invention. Antibodies of the present invention are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A- Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography. Therapeutic methods of the invention : The inventors have shown that cross-reacting F1M1 is able to reduce tumor growth without apparent toxicity in C57BL/6 mice harboring grafts of mouse E0771 cells (a strongly immune-infiltrated Cath-D-secreting basal-like TNBC cell line). The inventors have also demonstrated that F1M1-Fc+ inhibited growth of MDA-MB-231 cell xenografts, SUM159 cell xenografts, and of two TNBC-PDX (one resistant to neoadjuvant chemotherapy) without no apparent toxicity in nude mice. Moreover, in combination therapy, F1M1-Fc+ improved paclitaxel and enzalutamide therapeutic efficacy. Thus, the anti-Cathepsin-D antibody of the invention, or a fragment thereof or the immunoconjugates of the invention may be useful for treating any disease associated with Cathepsin-D overexpression preferentially cancers. The antibodies of the invention or a fragment thereof or the immunoconjugates of the invention may be used alone or in combination with any suitable agent. In another aspect, the invention relates to the anti-Cathepsin-D antibody of the invention, or a fragment thereof or the immunoconjugates of the invention, as described above, for use as a drug. Thus, the invention relates to an anti-Cathepsin-D antibody for use as a drug, wherein said antibody comprises : (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7 Thus, the invention relates to a human anti-Cathepsin-D antibody for use as a drug, wherein said antibody comprises: (a) a heavy chain wherein the variable domain has a sequence set forth as to SEQ ID NO:1, and (b) a light chain wherein the variable domain has a sequence set forth as to SEQ ID NO:5. In particular, the invention relates to an anti-Cathepsin-D antibody for use as a drug, wherein said antibody comprises: (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7, and (c) a FC region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T, I332E (“F1M1-Fc+”). In particular, the invention relates to an anti-Cathepsin-D antibody for use as a drug, wherein said antibody comprises: (a) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (b) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7, and (c) a Fc region comprising at least 1, 2 or 3 mutations selected from the group consisting of L234A, L235A and P329G (“F1M1-Fc-”). In some embodiments, the anti-Cathepsin-D antibody for use according to the invention is conjugated to a cytotoxic moiety. In a further embodiment, the anti-Cathepsin-D antibody or a fragment thereof or the immunoconjugates for use according to the invention in the treatment of hyperproliferative disorders or diseases. In other words, the invention relates to a method for treating hyperproliferative disorders or diseases in a subject in need thereof, comprising administering to said subject an effective amount of the anti-Cathepsin-D antibody of the invention or a fragment thereof. In another embodiment, the antibody or a fragment thereof for use in the treatment of hyperproliferative diseases. More particularly, the hyperproliferative diseases are associated with cath-D overexpression. As used herein, the term "abnormal cell growth" and "hyperproliferative disorders or diseases" are used interchangeably in this application and refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). In the context of the invention the hyperproliferative diseases refers to diseases having an overexpression of cathepsin-D. Typically, hyperproliferative diseases are selected but not limited to, cancer (e.g. breast cancer, renal cancer etc), skin disorders (e.g. psoriasis, wound healing), inflammatory diseases (e.g. inflammatory bowel disease). In a particular embodiment, the hyperproliferative disease is cancer. As used herein, the term “cancer” or “tumor” refers to a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body. The cancer that may treated by methods and compositions of the invention include, but are not limited to cancer cells from the bladder, blood, bone, bone marrow, brain, breast, colon, esophagus, gastrointestinal, gum, head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach, testis, tongue, or uterus. In addition, the cancer may specifically be of the following histological type, though it is not limited to these: neoplasm, malignant; carcinoma; carcinoma, undifferentiated; giant and spindle cell carcinoma; small cell carcinoma; papillary carcinoma; squamous cell carcinoma; lymphoepithelial carcinoma; basal cell carcinoma; pilomatrix carcinoma; transitional cell carcinoma; papillary transitional cell carcinoma; adenocarcinoma; gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma; adenoid cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma, familial polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchiolo-alveolar adenocarcinoma; papillary adenocarcinoma; chromophobe carcinoma; acidophil carcinoma; oxyphilic adenocarcinoma; basophil carcinoma; clear cell adenocarcinoma; granular cell carcinoma; follicular adenocarcinoma; papillary and follicular adenocarcinoma; nonencapsulating sclerosing carcinoma; adrenal cortical carcinoma; endometroid carcinoma; skin appendage carcinoma; apocrine adenocarcinoma; sebaceous adenocarcinoma; ceruminous; adenocarcinoma; mucoepidermoid carcinoma; cystadenocarcinoma; papillary cystadenocarcinoma; papillary serous cystadenocarcinoma; mucinous cystadenocarcinoma; mucinous adenocarcinoma; signet ring cell carcinoma; infiltrating duct carcinoma; medullary carcinoma; lobular carcinoma; inflammatory carcinoma; paget's disease, mammary; acinar cell carcinoma; adenosquamous carcinoma; adenocarcinoma w/squamous metaplasia; thymoma, malignant; ovarian stromal tumor, malignant; thecoma, malignant; granulosa cell tumor, malignant; and roblastoma, malignant; Sertoli cell carcinoma; leydig cell tumor, malignant; lipid cell tumor, malignant; paraganglioma, malignant; extra-mammary paraganglioma, malignant; pheochromocytoma; glomangiosarcoma; malignant melanoma; amelanotic melanoma; superficial spreading melanoma; malign melanoma in giant pigmented nevus; epithelioid cell melanoma; blue nevus, malignant; sarcoma; fibrosarcoma; fibrous histiocytoma, malignant; myxosarcoma; liposarcoma; leiomyosarcoma; rhabdomyosarcoma; embryonal rhabdomyosarcoma; alveolar rhabdomyosarcoma; stromal sarcoma; mixed tumor, malignant; mullerian mixed tumor; nephroblastoma; hepatoblastoma; carcinosarcoma; mesenchymoma, malignant; brenner tumor, malignant; phyllodes tumor, malignant; synovial sarcoma; mesothelioma, malignant; dysgerminoma; embryonal carcinoma; teratoma, malignant; struma ovarii, malignant; choriocarcinoma; mesonephroma, malignant; hemangiosarcoma; hemangioendothelioma, malignant; kaposi's sarcoma; hemangiopericytoma, malignant; lymphangiosarcoma; osteosarcoma; juxtacortical osteosarcoma; chondrosarcoma; chondroblastoma, malignant; mesenchymal chondrosarcoma; giant cell tumor of bone; ewing's sarcoma; odontogenic tumor, malignant; ameloblastic odontosarcoma; ameloblastoma, malignant; ameloblastic fibrosarcoma; pinealoma, malignant; chordoma; glioma, malignant; ependymoma; astrocytoma; protoplasmic astrocytoma; fibrillary astrocytoma; astroblastoma; glioblastoma; oligodendroglioma; oligodendroblastoma; primitive neuroectodermal; cerebellar sarcoma; ganglioneuroblastoma; neuroblastoma; retinoblastoma; olfactory neurogenic tumor; meningioma, malignant; neurofibrosarcoma; neurilemmoma, malignant; granular cell tumor, malignant; malignant lymphoma; Hodgkin's disease; Hodgkin's lymphoma; paragranuloma; malignant lymphoma, small lymphocytic; malignant lymphoma, large cell, diffuse; malignant lymphoma, follicular; mycosis fungoides; other specified non-Hodgkin's lymphomas; malignant histiocytosis; multiple myeloma; mast cell sarcoma; immunoproliferative small intestinal disease; leukemia; lymphoid leukemia; plasma cell leukemia; erythroleukemia; lymphosarcoma cell leukemia; myeloid leukemia; basophilic leukemia; eosinophilic leukemia; monocytic leukemia; mast cell leukemia; megakaryoblastic leukemia; myeloid sarcoma; and hairy cell leukemia. In a particular embodiment, the cancer is selected from the group consisting of breast cancer, melanoma, ovarian cancer, lung cancer, liver cancer, pancreatic cancer, melanoma, squamous cell carcinoma, endometrial cancer, head and neck cancer, bladder cancer, malignant glioma, prostate cancer, colon adenocarcinoma or gastric cancer. In a particular embodiment, the cancer exhibits an abundant stroma or tumoral microenvironment (“High stromal”, i.e at least 30% 40%, 50%, 60%, 70%, 80% and 90% of the tumor volume). As used herein, the term "stroma " or “microenvironment” has its general meaning in the art and refers to extracellular matrix and specialized connective tissue cells, including fibroblasts and mesenchymal stromal cells. Tumors have stroma and require stroma for nutritional support and the removal of waste products, but stromal content can vary markedly in different types of cancers. In the context of the invention, a high stromal refers to a tumors having a stroma representing at least 30% 40%, 50%, 60%, 70%, 80% and 90% of the tumor mass. In a particular embodiment, the cancer is an immune-inflamed tumor. As used herein, the term “Immune-inflamed tumors”, also named “hot tumors”, are characterized by high immune cell infiltration, especially T-cell infiltration, increased interferon-γ (IFN-γ) signaling and expression of PD-L1. In a particular embodiment, the cancer is a metastatic cancer. In a particular embodiment, the cancer is breast cancer or pancreatic cancer. In a particular embodiment, the breast cancer is an estrogen-receptor positive (ER+) hormono-resistant breast cancer or a triple-negative (ER- and PR-, HER2-non amplified) breast cancer (TNBC). In a particular embodiment, the breast cancer is TNBC. Some study has been made to classify TNBC into different subtypes, according to their molecular classification and in particular based to their tumor microenvironment (TME), as detailed in Zhao et al Molecular subtypes and precision treatment of triple-negative breast cancer. Ann Transl Med.2020 Apr;8(7):499. In a particular embodiment, the TNBC exhibit an abundant adaptive and innate immune cells infiltration (“immunity high TNBC”), as detailed in Zhao et al. In a particular embodiment, the TNBC is a LAR subtype characterized by androgen receptor signaling, as detailed in Zhao et al. In a particular embodiment, the TNBC is an IM subtype showing high immune cell signaling and cytokine signaling gene expression, as detailed in Zhao et al. In a particular embodiment, the cancer is a cancer wherein Cathepsin-D is overexpressed and hypersecreted in the tumor stroma or tumor microenvironment. The invention also relates to a method for treating a cancer wherein Cathepsin-D is overexpressed in a subject comprising administering to said subject an effective amount of the anti-Cathepsin-D antibody of the invention or a fragment thereof. The invention also relates to the anti-Cathepsin-D antibody of the invention or a fragment thereof for use in the treatment of cancer in a subject having a high level of Cathepsin- D. The above method and use comprise the steps of measuring the level of Cathepsin-D in a biological sample obtained from said subject and comparing this level to a reference control value. A high level of Cathepsin-D means that the antibody of the invention must be used. As used herein, the term “biological sample” refers to any sample obtained from a subject, such as a serum sample, a plasma sample, a urine sample, a blood sample, a lymph sample, or a tissue biopsy. In preferred embodiment, the biological sample is tumor tissue sample or stromal tissue sample. The level of the Cath-D may be determined by using standard electrophoretic and immunodiagnostic techniques, including immunoassays such as competition, direct reaction such as immunohistochemistry, or sandwich type assays. Such assays include, but are not limited to, Western blots; agglutination tests; enzyme-labelled and mediated immunoassays, such as ELISAs; biotin/avidin type assays; radioimmunoassays; immunoelectrophoresis; immunoprecipitation, etc. The reactions generally include revealing labels such as fluorescent, chemiluminescent, radioactive, enzymatic labels or dye molecules, or other methods for detecting the formation of a complex between the antigen and the antibody or antibodies reacted therewith. In a particular embodiment, the methods of the invention comprise contacting the biological sample with a binding partner. As used therein, binding partner refers to a molecule capable of selectively interacting with Cathepsin-D. The binding partner may be generally an antibody that may be polyclonal or monoclonal, preferably monoclonal. As used herein, the term “subject” refers to any mammals, such as a rodent, a feline, a canine, and a primate. Particularly, in the present invention, the subject is a human afflicted with or susceptible to be afflicted with a disease wherein Cathepsin-D is overexpressed. In another embodiment, the subject is a human afflicted with or susceptible to be afflicted with a cancer. In particular embodiment, the subject is a human afflicted with or susceptible to be afflicted with a cancer wherein Cathepsin-D is overexpressed, and in particularly breast cancer or pancreatic cancer wherein Cathepsin-D is overexpressed. In another embodiment, the subject is a human afflicted with or susceptible to be afflicted with TNBC. In some embodiment, the anti-Cathepsin-D antibody of the invention or a fragment thereof can be administered in combination with any suitable agent, in particular with anti- cancer therapy. As used herein, the term “anti-cancer therapy” has its general meaning in the art and refers to any compound, natural or synthetic, used for the treatment of cancer. In a particular embodiment, the classical treatment refers to radiation therapy, antibody therapy, immune checkpoint inhibitor, antiandrogens, CAR Therapy, such as CAR T- , CAR M- or CAR NK-cell therapy, antibody-drug conjugates (ADC) or chemotherapy. In some embodiment, the anti-Cathepsin-D antibody of the invention or a fragment thereof is administered in combination with an antibody-drug conjugates. Antibody-drug conjugates or ADCs are a class of biopharmaceutical drugs designed as a targeted therapy for treating cancer. Unlike chemotherapy, ADCs are intended to target and kill tumor cells while sparing healthy cells. ADC includes but are not limited to Gemtuzumab ozogamicin, Brentuximab vedotin, Trastuzumab emtansine, Inotuzumab ozogamicin, Polatuzumab vedotin, Enfortumab vedotin, Trastuzumab deruxtecan, Sacituzumab govitecan, Belantamab mafodotin, Moxetumomab pasudotox, Loncastuximab tesirine and Tisotumab vedotin-tftv. In some embodiment, the anti-Cathepsin-D antibody of the invention or a fragment thereof is administered in combination with a chemotherapeutic agent. In other words, in particular embodiment, the invention refers to i) the anti-Cathepsin- D antibody of the invention or a fragment thereof and ii) a chemotherapeutic agent as a combined preparation for simultaneous, separate or sequential use in the treatment of a cancer. As used herein, the term "chemotherapeutic agent" refers to chemical compounds that are effective in inhibiting tumor growth. Examples of chemotherapeutic agents include multkinase inhibitors such as sorafenib and sunitinib, alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaorarnide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a carnptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estrarnustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimus tine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as the enediyne antibiotics (e.g. calicheamicin, especially calicheamicin (11 and calicheamicin 211, see, e.g., Agnew Chem Intl. Ed. Engl. 33: 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6- diazo-5-oxo-L-norleucine, doxorubicin (including morpholino- doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idanrbicin, marcellomycin, mitomycins, mycophenolic acid, nogalarnycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptomgrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti- adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophospharnide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defo famine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pento statin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; razoxane; rhizoxin; sizofiran; spirogennanium; tenuazonic acid; triaziquone; 2,2',2"- trichlorotriethylarnine; trichothecenes (especially T-2 toxin, verracurin A, roridinA and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobromtol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g. paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and doxetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6- thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisp latin and carbop latin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-11 ; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included in this definition are antihormonal agents that act to regulate or inhibit honnone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. In particular embodiment, the chemotherapeutic agent is paclitaxel. In some embodiment, the anti-Cathepsin-D antibody of the invention is administered in combination with PROTAC or molecular glue degraders. In some embodiment, the anti-Cathepsin-D antibody of the invention is administered in combination with radiation therapy. In other words, in particular embodiment, the invention refers to i) the anti-Cathepsin- D antibody of the invention and ii) radiation therapy as a combined preparation for simultaneous, separate or sequential use in the treatment of a cancer. As used herein, the term “radiation therapy” has its general meaning in the art and refers the treatment of cancer with ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated (the target tissue) by damaging their genetic material, making it impossible for these cells to continue to grow. One type of radiation therapy commonly used involves photons, e.g. X-rays. Depending on the amount of energy they possess, the rays can be used to destroy cancer cells on the surface of or deeper in the body. The higher the energy of the x-ray beam, the deeper the x-rays can go into the target tissue. Linear accelerators and betatrons produce x-rays of increasingly greater energy. The use of machines to focus radiation (such as x-rays) on a cancer site is called external beam radiation therapy. Gamma rays are another form of photons used in radiation therapy. Gamma rays are produced spontaneously as certain elements (such as radium, uranium, and cobalt 60) release radiation as they decompose, or decay. In some embodiments, the radiation therapy is external radiation therapy. Examples of external radiation therapy include, but are not limited to, conventional external beam radiation therapy; three-dimensional conformal radiation therapy (3D-CRT), which delivers shaped beams to closely fit the shape of a tumor from different directions; intensity modulated radiation therapy (IMRT), e.g., helical tomotherapy, which shapes the radiation beams to closely fit the shape of a tumor and also alters the radiation dose according to the shape of the tumor; conformal proton beam radiation therapy; image-guided radiation therapy (IGRT), which combines scanning and radiation technologies to provide real time images of a tumor to guide the radiation treatment; intraoperative radiation therapy (IORT), which delivers radiation directly to a tumor during surgery; stereotactic radiosurgery, which delivers a large, precise radiation dose to a small tumor area in a single session; hyperfractionated radiation therapy, e.g., continuous hyperfractionated accelerated radiation therapy (CHART), in which more than one treatment (fraction) of radiation therapy are given to a subject per day; and hypofractionated radiation therapy, in which larger doses of radiation therapy per fraction is given but fewer fractions. In some embodiment, the anti-Cathepsin-D antibody of the invention or a fragment thereof is administered in combination with an immune checkpoint inhibitor. In other words, in particular embodiment, the invention refers to i) the anti-Cathepsin- D antibody of the invention or a fragment thereof and ii) an immune checkpoint inhibitor as a combined preparation for simultaneous, separate or sequential use in the treatment of a cancer. As used herein, the term "immune checkpoint protein" has its general meaning in the art and refers to a molecule that is expressed by T lymphocytes in that either turn up a signal (stimulatory checkpoint molecules) or turn down a signal (inhibitory checkpoint molecules). Immune checkpoints are the regulators of the immune system. They are crucial for self- tolerance, which prevents the immune system from attacking cells indiscriminately. Immune checkpoints are targets for cancer immunotherapy due to their potential for use in multiple types of cancers. Typically, by using immune checkpoint inhibitors, the anti-tumoral response is reactivated by reactivation of cytotoxic T- lymphocytes. The anti-Cathepsin-D antibody antibody of the invention as described above can be combined with an immune checkpoint inhibitor to inhibit the recruitment of immunosuppressive tumor-associated macrophages M2 and myeloid-derived suppressor cells. Immune checkpoint molecules are recognized in the art to constitute immune checkpoint pathways similar to the CTLA-4 and PD-1 dependent pathways (see e.g. Pardoll, 2012. Nature Rev Cancer 12:252-264; Mellman et al. , 2011. Nature 480:480- 489). Examples of stimulatory checkpoint molecules include CD27, CD28, CD40, CD122, CD137, OX40, GITR, and ICOS. Examples of inhibitory checkpoint molecules include A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 and VISTA. The Adenosine A2A receptor (A2AR) is regarded as an important checkpoint in cancer therapy because adenosine in the immune microenvironment, leading to the activation of the A2a receptor, is negative immune feedback loop and the tumor microenvironment has relatively high concentrations of adenosine. B7-H3, also called CD276, was originally understood to be a co-stimulatory molecule but is now regarded as co-inhibitory. B7-H4, also called VTCN1, is expressed by tumor cells and tumor- associated macrophages and plays a role in tumour escape. B and T Lymphocyte Attenuator (BTLA) and also called CD272, has HVEM (Herpesvirus Entry Mediator) as its ligand. Surface expression of BTLA is gradually downregulated during differentiation of human CD8+ T cells from the naive to effector cell phenotype, however tumor-specific human CD8+ T cells express high levels of BTLA. CTLA-4, Cytotoxic T-Lymphocyte-Associated protein 4 and also called CD152. Expression of CTLA-4 on Treg cells serves to control T cell proliferation. IDO, Indoleamine 2,3-dioxygenase, is a tryptophan catabolic enzyme. A related immune-inhibitory enzymes. Another important molecule is TDO, tryptophan 2,3-dioxygenase. IDO is known to suppress T and NK cells, generate and activate Tregs and myeloid-derived suppressor cells, and promote tumour angiogenesis. KIR, Killer-cell Immunoglobulin-like Receptor, is a receptor for MHC Class I molecules on Natural Killer cells. LAG3, Lymphocyte Activation Gene-3, works to suppress an immune response by action to Tregs as well as direct effects on CD8+ T cells. PD-1, Programmed Death 1 (PD-1) receptor, has two ligands, PD-L1 and PD-L2. This checkpoint is the target of Merck & Co.'s melanoma drug Keytruda, which gained FDA approval in September 2014. An advantage of targeting PD-1 is that it can restore immune function in the tumor microenvironment. TIM-3, short for T-cell Immunoglobulin domain and Mucin domain 3, expresses on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. TIM-3 acts as a negative regulator of Th1/Tc1 function by triggering cell death upon interaction with its ligand, galectin-9. VISTA, Short for V-domain Ig suppressor of T cell activation, VISTA is primarily expressed on hematopoietic cells so that consistent expression of VISTA on leukocytes within tumors may allow VISTA blockade to be effective across a broad range of solid tumors. Tumor cells often take advantage of these checkpoints to escape detection by the immune system. Thus, inhibiting a checkpoint protein on the immune system may enhance the anti-tumor T-cell response. In some embodiments, an immune checkpoint inhibitor refers to any compound inhibiting the function of an immune checkpoint protein. Inhibition includes reduction of function and full blockade. In some embodiments, the immune checkpoint inhibitor could be an antibody, synthetic or native sequence peptides, small molecules or aptamers which bind to the immune checkpoint proteins and their ligands. In a particular embodiment, the immune checkpoint inhibitor is an antibody. Typically, antibodies are directed against A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA. In a particular embodiment, the immune checkpoint inhibitor is an anti-PD-1 antibody such as Pembrolizumab (Keytruda), Nivolumab (Opdivo) and Cemiplimab (Libtayo). In some embodiments, the immune checkpoint inhibitor is an anti-PD-L1 antibody such as Atezolizumab (Tecentriq), Durvalumab (Imfinzi), Avelumab and BMS-936559 (BMS). In some embodiments, the immune checkpoint inhibitor is an anti-PD-L2 antibody such as described in US7709214, US7432059 and US8552154. In some embodiments, the immune checkpoint inhibitor is an anti-Tim-3 antibody such as described in WO03063792, WO2011155607, WO2015117002, WO2010117057 and WO2013006490. In some embodiments, the immune checkpoint inhibitor is an anti-CTLA-4 antibody such as Ipilimumab (Yervoy) and tremelimumab (Imjuno). In some embodiments, the immune checkpoint inhibitor is an anti-LAG-3 antibody such as Relatlimab. In some embodiments, the immune checkpoint inhibitor is a small organic molecule. The term "small organic molecule" as used herein, refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes biological macro molecules (e. g. proteins, nucleic acids, etc.). Typically, small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da. Typically, the small organic molecules interfere with transduction pathway of A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA. In a particular embodiment, small organic molecules interfere with transduction pathway of PD-1 and Tim-3. For example, they can interfere with molecules, receptors or enzymes involved in PD-1 and Tim-3 pathway. In a particular embodiment, the small organic molecules interfere with Indoleamine- pyrrole 2,3-dioxygenase (IDO) inhibitor. IDO is involved in the tryptophan catabolism (Liu et al 2010, Vacchelli et al 2014, Zhai et al 2015). Examples of IDO inhibitors are described in WO 2014150677. Examples of IDO inhibitors include without limitation 1-methyl-tryptoρhan (IMT), β- (3-benzofuranyl)-alanine, β-(3-benzo(b)thienyl)-alanine), 6-nitro-tryptophan, 6- fluoro-tryptophan, 4-methyl-tryptophan, 5 -methyl tryptophan, 6-methyl-tryptophan, 5- methoxy-tryptophan, 5 -hydroxy-tryptophan, indole 3-carbinol, 3,3'- diindolylmethane, epigallocatechin gallate, 5-Br-4-Cl-indoxyl 1,3-diacetate, 9- vinylcarbazole, acemetacin, 5- bromo-tryptophan, 5-bromoindoxyl diacetate, 3- Amino-naphtoic acid, pyrrolidine dithiocarbamate, 4-phenylimidazole a brassinin derivative, a thiohydantoin derivative, a β- carboline derivative or a brassilexin derivative. In a particular embodiment, the IDO inhibitor is selected from 1-methyl-tryptophan, β-(3- benzofuranyl)-alanine, 6-nitro-L-tryptophan, 3- Amino-naphtoic acid and β-[3- benzo(b)thienyl] -alanine or a derivative or prodrug thereof. In a particular embodiment, the inhibitor of IDO is Epacadostat, (INCB24360, INCB024360) has the following chemical formula in the art and refers to -N-(3-bromo-4- fluorophényl)-N'-hydroxy-4-{[2-(sulfamoylamino)-éthyl]amino}-1,2,5-oxadiazole-3 carboximidamide : In a particular embodiment, the inhibitor is BGB324, also called R428, such as described in WO2009054864, refers to 1H-1,2,4-Triazole-3,5-diamine, 1-(6,7-dihydro-5H- benzo[6,7]cyclohepta[1,2-c]pyridazin-3-yl)-N3-[(7S)-6,7,8,9-tetrahydro-7-(1-pyrrolidinyl)- 5H-benzocyclohepten-2-yl]- and has the following formula in the art: In a particular embodiment, the inhibitor is CA-170 (or AUPM-170): an oral, small molecule immune checkpoint antagonist targeting programmed death ligand-1 (PD-L1) and V- domain Ig suppressor of T cell activation (VISTA) (Liu et al 2015). Preclinical data of CA-170 are presented by Curis Collaborator and Aurigene on November at ACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics. In some embodiments, the immune checkpoint inhibitor is an aptamer. Typically, the aptamers are directed against A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA. In a particular embodiment, aptamers are DNA aptamers such as described in Prodeus et al 2015. A major disadvantage of aptamers as therapeutic entities is their poor pharmacokinetic profiles, as these short DNA strands are rapidly removed from circulation due to renal filtration. Thus, aptamers according to the invention are conjugated to with high molecular weight polymers such as polyethylene glycol (PEG). In a particular embodiment, the aptamer is an anti-PD-1 aptamer. Particularly, the anti-PD-1 aptamer is MP7 pegylated as described in Prodeus et al 2015. In some embodiments, the immune check point inhibitor is selected from the group consisting of PD-1 inhibitors such as Pembrolizumab (Keytruda), Nivolumab (Opdivo), Cemiplimab (Libtayo), PD-L1 inhibitors such as Atezolizumab (Tecentriq), Avelumab (Bavencio), Durvalumab (Imfinzi), CTLA-4 inhibitors such as Ipilimumab (Yervoy) and tremelimumab (Imjuno) and LAG-3 inhibitors such as Relatlimab. Thus, in particular embodiment, the invention refers to i) the human anti-Cathepsin-D antibody of the invention and ii) an immune checkpoint inhibitor as a combined preparation for simultaneous, separate or sequential use in the treatment of a cancer, wherein the immune check point inhibitor is selected from the group consisting of PD-1 inhibitors such as Pembrolizumab (Keytruda), Nivolumab (Opdivo), Cemiplimab (Libtayo), PD-L1 inhibitors such as Atezolizumab (Tecentriq), Avelumab (Bavencio), Durvalumab (Imfinzi), CTLA-4 inhibitors such as Ipilimumab (Yervoy) and tremelimumab (Imjuno) and LAG-3 inhibitors such as Relatlimab. In some embodiment, the anti-Cathepsin-D antibody of the invention or a fragment thereof is administered in combination with an antiandrogens. In other words, in particular embodiment, the invention refers to i) the anti-Cathepsin- D antibody of the invention or a fragment thereof and ii) an antiandrogens as a combined preparation for simultaneous, separate or sequential use in the treatment of a cancer. As used herein, the term "antiandrogens" has its general meaning in the art and refers to a molecule that prevent androgens like testosterone and dihydrotestosterone (DHT) from mediating their biological effects in the body. They act by blocking the androgen receptor (AR) and/or inhibiting or suppressing androgen production. Examples of antiandrogens include but are not limited to : androgen receptor antagonists such as steroidal antiandrogens cyproterone acetate, megestrol acetate, chlormadinone acetate, spironolactone, oxendolone ; nonsteroidal antiandrogens flutamide, bicalutamide, nilutamide, topilutamide, enzalutamide, and apalutamide, aside from cyproterone acetate and chlormadinone acetate, dienogest, drospirenone, medrogestone, nomegestrol acetate, promegestone, and trimegestone ; androgen synthesis inhibitors such as CYP17A1 inhibitors ketoconazole, abiraterone acetate, and seviteronel, the CYP11A1 (P450scc) inhibitor aminoglutethimide, and the 5α-reductase inhibitors finasteride, dutasteride, epristeride, alfatradiol, and saw palmetto extract (Serenoa repens), cyproterone acetate, spironolactone, medrogestone, flutamide, nilutamide, and bifluranol ; antigonadotropins such as GnRH modulators like leuprorelin, progestogens like allylestrenol, chlormadinone acetate, cyproterone acetate, gestonorone caproate, hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, osaterone acetate (veterinary), and oxendolone, and estrogens like estradiol, estradiol esters, ethinylestradiol, conjugated estrogens, and diethylstilbestrol. In particular embodiment, the antiandrogen is enzalutamide. The compounds used in combination with the human anti- Cathepsin-D antibody of the present invention are administered and dosed in accordance with good medical practice, taking into account the clinical condition of the individual subject, the site and method of administration, scheduling of administration, patient age, sex, body weight and other factors known to medical practitioners. The pharmaceutically “effective amount” for purposes herein is thus determined by such considerations as are known in the art. The amount must be effective to achieve improvement including, but not limited to, improved survival rate or more rapid recovery, or improvement or elimination of symptoms and other indicators as are selected as appropriate measures by those skilled in the art. Pharmaceutical compositions of the invention: The anti-Cathepsin-D antibody of the invention or a fragment thereof as described above may be combined with pharmaceutically acceptable excipients, and optionally sustained- release matrices, such as biodegradable polymers, to form therapeutic compositions. Accordingly, the present invention relates to a pharmaceutical composition comprising an anti-Cathepsin-D antibody according to the invention or a fragment thereof or a immunoconjugate of the invention and a pharmaceutically acceptable carrier. The present invention also relates to a pharmaceutical composition for use as drug, wherein said pharmaceutical composition comprises an anti-Cathepsin-D antibody according to the invention or a fragment thereof or a immunoconjugate of the invention and a pharmaceutically acceptable carrier. The present invention also relates to a pharmaceutical composition for use in the treatment of hyperproliferative disorders or diseases in a subject in need thereof, wherein said pharmaceutical composition comprises an anti-Cathepsin-D antibody or a fragment thereof according to the invention and a pharmaceutically acceptable carrier. In particular embodiment, the hyperproliferative disorders or diseases is cancer, and more particularly breast cancer. Indeed, the antibody for use according to the invention alone and/or combined with any suitable agent as described above may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form pharmaceutical compositions. "Pharmaceutically" or "pharmaceutically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate. A pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, the active principle, alone or in combination with another active principle, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings. Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms. Typically, the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The polypeptide (or nucleic acid encoding thereof) can be formulated into a composition in a neutral or salt form. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. The carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active polypeptides in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum- drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention. FIGURES: Figure 1: Binding of F1 and F1M1 to Cath-D secreted from E0771 TNBC mouse cell line. Sandwich ELISA in which secreted Cath-D from E0771 cell (left panel) supernatants was added to wells pre-coated with an anti-Cath-D polyclonal antibody (#AF1029) in the presence of increasing concentrations of F1 and F1M1 anti-Cath-D antibodies. Binding of F1 and F1M1 to secreted Cath-D was revealed with an anti-mouse F(ab’)2 HRP conjugated antibody. Mean±SD (n=3). The EC50 values are shown. Similar results were obtained in 5 independent experiments. Figure 2: Therapeutic effect of the F1 and F1M1 anti-Cath-D mouse IgG2a antibodies in immunocompetent C57BL/6 mice grafted with TNBC cell line E0771. (A). Tumor growth in mice grafted with E0771 cells. At day 2 post graft, C57BL/6 mice were treated with F1 (n=9), F1M1 (n=9) or C1.18.4 isotype control (CTRL n= 9) (15 mg/kg) was initiated (intraperitoneal injection three times per week for 33 days). All mice were sacrificed at day 33. Tumor volume (in mm3) is shown as the mean ±SEM. of 9 mice per group. *, P<0.05, significantly different as indicated. **P=0.005 for F1 versus CTRL, ** P=0.002 for F1M1 versus CTRL (mixed-effects multiple linear regression test). (B) Individual tumor growth curves in mice grafted with E0771 cells. Spider plots show the tumor growth of each mouse in each group CTRL (n=9, left panel), and F1M1 (n=9, right panel). (C) Mean tumor volume at day 33 in mice grafted with E0771 cells. n= 9 for CTRL; n= 9 for F1; n=9 for F1M1. *, P<0.05, significantly different as indicated. *P=0.0136 for F1 versus CTRL, *P=0.0194 for F1M1 versus CTRL (Mann Whitney t test); data are the mean ±SEM of 9 mice per group. (D) Weight monitoring in mice grafted with E0771 cells. n= 9 for CTRL; n=9 for F1and n= 9 for F1M1, data are the mean ±SEM of 9 mice per group. Figure 3. Anti-Cath-D antibody-based therapy rebalances TAM recruitment and activation in E0771 cell-derived tumors. (A) TAM recruitment. The percentage of F4/80+ CD11b+ TAMs was quantified by FACS and expressed relative to all CD45+ immune cells in the tumor (n=9 for CTRL; n=7 for F1; n=8 for F1M1); *, P<0.05, significantly different as indicated. *P= 0.0393 for F1 versus CTRL, *P=0.0464 for F1M1 versus C1.18.1 control (CTRL) (Mann-Whitney t-test); mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. (B) M2-TAM recruitment. The percentage of F4/80+ CD11b+ CD206+ M2-TAMs (left panel) was quantified by FACS and expressed relative to all CD45+ immune cells in the tumor (n=9 for CTRL; n=7 for F1; n=8 for F1M1); *, P<0.05, significantly different as indicated. *P= 0.0311 for F1 versus CTRL, *P=0.0111 for F1M1 versus CTRL (Mann-Whitney t-test); data are the mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. Linear regression analysis of M2-TAMs and tumor volumes (right panel): R2=0.1988, *P=0.029, n=24. (C) M1-TAM recruitment and activation. The percentage of F4/80+ CD11b+ CD11c+ CD206- M1-TAMs (left panel) was quantified by FACS and expressed relative to all CD45+ immune cells in the tumor (n=9 for CTRL; n=7 for F1; n=8 for F1M1); P= 0.8371 for F1 versus CTRL, P=0.8148 for F1M1 versus CTRL (Mann-Whitney t-test); data are the mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. The percentage of activated CD86+ M1-TAMs (middle panel) was quantified by FACS and expressed relative to all M1-TAMs (n=9 for CTRL; n=7 for F1; n=8 for F1M1), *, P<0.05, significantly different as indicated. P= 0.14 for F1 versus CTRL; *P=0.0495 for F1M1 versus CTRL (Mann-Whitney t-test); mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. Linear regression analysis of CD86+ M1-TAMs and tumor volumes (right panel): R2=0.2087, *P=0.0248, n=24. (D) Quantification of Ccl2, Ccr2, Il10 and Mmp9 mRNA expression. Total RNA was extracted from E0771 cell-derived tumors at treatment end (day 33) and Ccl2, Ccr2, Il10 and Mmp9 expression levels were analyzed by RT- qPCR. Data are the mean ± SEM relative to Rps9 expression (CTRL n=8; F1 n=5; F1M1 n=7). For Ccl2, *P=0.0575 for F1 versus CTRL and *P=0.0190 for F1M1 versus CTRL. For Ccr2, **P=0.0016 for F1 versus CTRL and *P=0.0185 for F1M1 versus CTRL. For Il10, **P=0.0031 for F1 versus CTRL and ***P=0.0003 for F1M1 versus CTRL. For Mmp9, **P=0.0031 for F1 versus CTRL and ***P=0.0003 for F1M1 versus CTRL (Mann-Whitney t-test). *, P<0.05, significantly different as indicated. Figure 4. Anti-Cath-D antibody-based therapy triggers NK cell activation in E0771 cell-derived tumors. (A) NK cell recruitment and activation. The percentage of NKp46+ NK cells was quantified by FACS and expressed relative to all CD45+ immune cells in the tumor (n=9 for CTRL; n=7 for F1; n=8 for F1M1) (left panel). P=0.3510 for F1 vs CTRL, P=0.8884 for F1M1 vs CTRL. The percentage of activated (CD16+) NK cells was quantified by FACS and expressed relative to all NK cells in the tumor (n=9 for CTRL; n=7 for F1; n=8 for F1M1) (right panel); *, P<0.05, significantly different as indicated. ***P=0.0002 for F1 versus CTRL, *P=0.0142 for F1M1 versus CTRL (Mann-Whitney t-test); data are the mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. (B) Quantification of Il15 and Tnf mRNA expression. Total RNA was extracted from E0771 cell-derived tumors at treatment end (day 33), and Il15 and Tnf were analyzed by RT-qPCR. Data are the mean ± SEM relative to RPS9 expression (CTRL n=8; F1 n=5; F1M1 n=7). For Il15, *P=0.0109 for F1 versus CTRL and P=0.0939 for F1M1 versus CTRL. For Tnf, *P=0.0295 for F1 versus CTRL and ***P=0.0003 for F1M1 versus CTRL (Mann-Whitney t-test). Figure 5. Anti-Cath-D antibody-based therapy induces the recruitment and maturation of cDC1 cells in E0771 cell-derived tumors. (A) cDC recruitment. The percentage of the cDC (left panel), cDC2 (middle panel) and cDC1 (right panel) subtypes was quantified by FACS and expressed relative to all CD45+ immune cells in the tumor (n=9 for CTRL; n=7 for F1; n=8 for F1M1). *, P<0.05, significantly different as indicated. For CD11c+/hi cDC, P=0.2991 for F1 versus CTRL and; P=0.5414 for F1M1 versus CTRL. For CD11c+/hi CD8- CD11b+ cDC2 cells, P>0.9 for F1 versus CTRL and P=0.6058 for F1M1 versus CTRL. For CD11c+/hi CD8+ CD11b- cDC1 cells, *P=0.0164 for F1 versus CTRL and **P=0.0055 for F1M1 versus CTRL (Mann-Whitney t-test); data are the mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. (B) cDC1 cell activation. The percentage of activated CD86+ cDC1 cells was quantified by FACS and expressed relative to all cDC1 cells in the tumor (n=9 for CTRL; n=7 for F1; n=8 for F1M1). *, P<0.05, significantly different as indicated. *P=0.0360 for F1 versus CTRL, **P=0.0053 for F1M1 versus CTRL (Mann-Whitney t-test); data are the mean ± SEM 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. (C) Quantification of H2d1, H2k1, B2m, and Il12p40 mRNA expression. Total RNA was extracted from E0771 cell-derived tumors at treatment end (day 33). The expression levels of H2d1, H2k1, B2m, and Il12p40 were quantified by RT-qPCR. Data are the mean ± SEM relative to Rps9 expression (CTRL n=8; F1 n=5; F1M1 n=7). For H2d1, **P=0.0016 for F1 versus CTRL and ***P=0.0003 for F1M1 versus CTRL. *, P<0.05, significantly different as indicated. For H2k1, **P=0.0016 for F1 versus CTRL and ***P=0.0003 for F1M1 versus CTRL. For B2m, **P=0.0016 for F1 versus CTRL and ***P=0.0003 for F1M1 versus CTRL. For Il12p40, P=0.0870 for F1 versus CTRL and **P= 0.0022 for F1M1 versus CTRL (Mann-Whitney t-test). Figure 6. Anti-Cath-D antibody-based therapy reduces T-cell exhaustion in E0771 cell-derived tumors. (A) Recruitment of CD4+ T cell and PDL-1+ CD4+ T cells. The percentage of CD4+ T cells was quantified by FACS and expressed relative to all CD45+ immune cells in the tumor (left panel). P=0.9182 for F1 versus CTRL and P=0.6730 for F1M1 versus CTRL. The percentage of PDL1+ CD4+ T cells was quantified by FACS and expressed relative to all CD4+ T cells in the tumor (right panel). *P=0.0164 for F1 versus CTRL and *P=0.0206 for F1M1 versus CTRL (Mann-Whitney t-test); data are the mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. *, P<0.05, significantly different as indicated. (B) Recruitment of CD8+ T cells and PDL-1+ CD8+ T cells. The percentage of CD8+ T cells was quantified by FACS and expressed relative to all CD45+ immune cells in the tumor (left panel). P=0.6065 for F1 versus CTRL and P=0.4807 for F1M1 versus CTRL. The percentage of PDL1+ CD8+ T cells was quantified by FACS and expressed relative to all CD8+ T cells in the tumor (right panel). *P=0.0311 for F1 versus CTRL and *P=0.036 for F1M1 versus CTRL (Mann-Whitney t-test); data are the mean ± SEM of 9 mice for CTRL, 7 mice for F1, and 8 mice for F1M1. *, P<0.05, significantly different as indicated. (C) Quantification of Lag3 and Tigit mRNA expression. Total RNA was extracted from E0771 cell-derived tumors at treatment end (day 33) and Lag3 (left panel) and Tigit (right panel) were analyzed by RT-qPCR. Data are the mean ± SEM relative to Rps9 expression (CTRL n=8; F1 n=5; F1M1 n=7). *, P<0.05, significantly different as indicated. For Lag3, *P=0.0109 for F1 versus CTRL and ***P=0.0006 for F1M1 versus CTRL. For Tigit, **P=0.0016 for F1 versus CTRL and **P=0.0012 for F1M1 versus CTRL (Mann-Whitney t-test). (D) Quantification of Foxp3 and IL4 mRNA expression. Total RNA was extracted and analyzed by RT-qPCR. Data are the mean ± SEM relative to Rps9 expression (CTRL n=8; F1 n=5; F1M1 n=7). *, P<0.05, significantly different as indicated. For Foxp3, **P=0.0016 for F1 versus CTRL and *P=0.0129 for F1M1 versus CTRL. For Il4, **P=0.0016 for F1 versus CTRL and *P=0.0180 for F1M1 versus CTRL (Mann-Whitney t-test). Figure 7. Cath-D is a tumor microenvironment antigen eligible to trigger ADCC and relevant for Fc-engineered antibody-based targeted therapy in TNBC (A) Binding of human IgG1 F1 and F1M1 to mouse Cath-D secreted from E0771Luc cells. Sandwich ELISA in which mouse cath-D from conditioned medium of E0771Luc cells was added to wells pre-coated with the anti-mouse cath-D AF1029 antibody in the presence of increasing concentrations of the IgG1 F1 or F1M1. Binding of F1 and F1M1 to secreted mouse cath-D was revealed with an anti-mouse F(ab’)2 HRP-conjugated antibody. The EC50 values are shown. (B) Binding of human IgG1 F1M1, F1M1-Fc-, and F1M1-Fc+ to human pro-cath-D secreted from MDA-MB-231 cells. Sandwich ELISA in which pro-cath- D from conditioned medium of MDA-MB-231 cells was added to wells pre-coated with the anti-human pro-cath-D M2E8 monoclonal antibody in the presence of increasing concentrations of F1M1, F1M1-Fc-, or F1M1-Fc+. Binding of F1M1, F1M1-Fc- and F1M1-Fc+ to pro-cath-D was revealed with an anti-human Fc HRP-conjugated antibody. The EC50 values are shown. (C) Binding of F1M1, F1M1-Fc-, and F1M1-Fc+ to human CD16a 158V and CD16a 158F by surface plasmon resonance. Increasing concentrations of F1M1, F1M1-Fc-, and F1M1-Fc+ (3.7, 11, 33, 100, 300 nM) were injected into the sensor chip on which hCD16a 158V (left panel) or hCD16a 158F (right panel) had been captured. The KD values are shown. (D) Binding of F1M1, F1M1-Fc-, and F1M1-Fc+ to human CD16a 158V and CD16a 158F by flow cytometry. NK92 cells transduced to express hCD16a 158V (left panel) or hCD16a 158F (right panel) were incubated with increasing concentrations (0.6 to 6666 nM) of F1M1- Fc-, F1M1 or F1M1-Fc+ at 4°C for 30min, followed by incubation with the APC-AF750 conjugated anti-CD163G8 antibody (positive control of human CD16 engagement) at 4°C for 30min. Results were expressed as percentage of fluorescent 3G8 binding: (MFI without anti- cath-D antibody - MFI with anti-cath-D antibody) × 100/ (MFI without anti-cath-D antibody). This allowed the standardization and comparison of results between NK92 hCD16a 158F and NK92 hCD16a 158V. Figure 8. Fc-engineered F1M1-Fc+ increases NK cell activation in vitro, and ADCC in TNBC cells and CAFs (A) Functional responses of NK92 hCD16a 158V and NK92 hCD16a 158F cells induced by F1M1-Fc-, F1M1, and F1M1-Fc+. For in vitro stimulation and functional responses of hCD16a-transduced NK92 cells, 96-well plates were sensitized with a saturating concentration (10 µg/ml; 66.6nM) of F1M1-Fc-, F1M1, F1M1-Fc+, or anti-hCD16a (3G8) monoclonal antibodies at 4°C for 12h. NK92 hCD16a 158V and NK92 hCD16a 158F cells were incubated on non-sensitized or sensitized plates with/without the anti-CD107a-PC5 antibody (1:20) and 0.1 µg/ml BD GolgiPlug containing brefeldin A at 37°C for 4h. Cells were then fixed and permeabilized and stained with an anti-IFNγ-PE antibody (1:20) at 4°C for 30min to detect intracellular IFNγ. The percentages of degranulating NK92 cells (CD107a+IFNγ-; white bars), IFNγ-producing NK92 cells (CD107a-IFNγ+; black bars), and NK92 cells exhibiting both responses (CD107a+IFNγ+; gray bars) were quantified by flow cytometry after 4h of stimulation. Mean (%) ± SD (n=3).3G8 (anti-hCD16a antibody) was used as positive control of CD16 engagement (Ctrl+). (B) Functional responses of NK92-hCD16a 158V cells induced by F1M1-Fc+ in the presence of increasing concentrations of cath-D. 96-well plates were coated with the M2E8 anti-cath-D antibody (2500 ng/well) at 4°C overnight, incubated or not with increasing concentrations of cath-D (3-50 nM) and then sensitized with F1M1-Fc+ (10 µg/ml). The percentage of degranulating (CD107a+IFNγ; white bars), IFNγ-producing (CD107a-IFNγ+; black bars), and NK92-hCD16a 158V cells exhibiting both responses (CD107a+IFNγ+; gray bars) was evaluated as in (A) after 4h of stimulation. (C) ADCC activity against MDA-MB-231 cells in the presence of NK92-hCD16a cells in response to F1M1-Fc-, F1M1 or F1M1-Fc+. MDA-MB-231 cells (target) were pre-incubated with F1M1-Fc-, F1M1, F1M1-Fc+, or with cetuximab at 100 µg/ml (666 nM) at 37°C for 30min, followed by incubation with NK92-hCD16a 158V (left) or NK92-hCD16a 158F (right) cells (effector) at an effector:target ratio of 10:1 for 24h. MDA-MB-231 cell lysis was evaluated by measuring LDH release by bioluminescence. Cetuximab (Cetux; anti-EGFR antibody) was used as positive control (Ctrl+). For hCD16a 158V cells (left): ****P <0.0001 for F1M1-Fc+ versus F1M1, ****P <0.0001 for F1M1-Fc+ versus F1M1-Fc-, *** P=0.0003 for F1M1 versus F1M1-Fc- (one-way ANOVA). For hCD16a 158F cells (right): ***P=0.0002 for F1M1-Fc+ versus F1M1, ****P <0.0001 for F1M1-Fc+ versus F1M1-Fc-, *P=0.015 for F1M1 versus F1M1-Fc- (one-way ANOVA). (D) Dose-dependent induction of ADCC against MDA-MB- 231 cells in the presence of hCD16a 158V-expressing NK92 cells in response to F1M1-Fc+. ADCC was evaluated in MDA-MB-231 cells incubated with hCD16a 158V-expressing NK92 cells at an effector:target ratio of 10:1, as described in (C), after incubation with increasing concentrations of F1M1-Fc+, from 0.1 to 666nM (0.015-100µg/ml). (E) Effect of blocking Fc binding sites in hCD16a on F1M1-Fc+-induced ADCC in MDA-MB-231 cells. NK92- hCD16a 158V cells were pre-incubated or not with Fc block (to saturate the Fc binding sites in hCD16a) for 30min. MDA-MB-231 cells, pre-incubated with F1M1-Fc+ at 100 µg/ml (666 nM) for 30min, were incubated with NK92-hCD16a 158V cells pre-incubated or not with Fc block. ADCC was analyzed as described in (C). Cetux, cetuximab as positive control (100µg/ml); F1M1-Fc-, negative control (100µg/ml). **P=0.0023 for F1M1-Fc+ versus F1M1-Fc++Fc block, **P=0.0012 for F1M1-Fc+ versus F1M1-Fc- (one-way ANOVA). (F) Blocking cath-D binding to Mannose-6-phosphate (M6P) receptors in MDA-MB-231 cells affects F1M1- Fc+-induced ADCC in the presence of NK92-hCD16a 158V cells. MDA-MB-231 cells (target) were pre-incubated with M6P (10 mM) or G6P (10 mM) for 24h. Then, ADCC was assessed in these cells with NK92-hCD16a 158V cells (effector) at an effector:target ratio of 10:1, as described in (C), after incubation with F1M1-Fc+ at 100µg/ml (666nM) in the presence of M6P (10 mM) or G6P (10 mM). M6P, mannose-6-phosphate; G6P, glucose-6-phosphate (negative control for M6P). Cetux, positive control (100µg/ml); F1M1-Fc-, negative control (100µg/ml). ****P <0.0001 for F1M1-Fc+ versus F1M1-Fc++M6P, ***P=0.0002 for F1M1- Fc++G6P versus F1M1-Fc++M6P (one-way ANOVA). (G) Blocking cath-D binding to M6P receptors in hCAFs affects F1M1-Fc+-induced ADCC in the presence of NK92-hCD16a 158V cells. Breast hCAF1 cells (target) were pre-incubated with M6P (10 mM) or G6P (10 mM) for 24h. ADCC was assessed in these hCAF1 cells in the presence of NK92-hCD16a 158V cells (effector) at an effector:target ratio of 10:1, as described in (F), after incubation with F1M1-Fc+ at 100µg/ml (666nM) and M6P (10 mM) or G6P (10 mM). Cetux, cetuximab as positive control (100µg/ml); F1M1-Fc-, negative control (100µg/ml). ****P <0.0001 for F1M1-Fc+ versus F1M1-Fc++M6P, ****P <0.0001 for F1M1-Fc++G6P versus F1M1- Fc++M6P (one-way ANOVA). (H) ADCC against MDA-MB-231 cell spheroids in the presence of NK92-hCD16a 158V cells in response to F1M1-Fc+. MDA-MB-231 cell spheroids (target) were first incubated with F1M1-Fc+, F1M1-Fc-, or cetuximab (Cetux) at 37 °C for 30min. Then, spheroids were incubated with NK92-hCD16a 158V cells (effector) at an effector:target ratio of 20:1 for 24h, and ADCC was assessed as described in (C). ****P <0.0001 for F1M1-Fc+ versus F1M1-Fc- (one-way ANOVA). Figure 9. F1M1-Fc+ is the best candidate to reduce growth of MDA-MB-231 cell xenografts and improve mouse survival. (A) Tumor growth. MDA-MB-231 cells were subcutaneously injected in nude mice. When tumor volume reached 50 mm3, mice were treated with F1M1 (n=10), F1M1-Fc- (n=8), F1M1-Fc+ (n=9) (15mg/kg), or rituximab (Ctrl; n=7) (15mg/kg), three times per week for 35 days. Mice were sacrificed when tumor volume reached 2000 mm3. Tumor volume (in mm3) is shown as the mean ± SEM. P=0.057 for F1M1-Fc- versus Ctrl, **P=0.011 for F1M1 versus Ctrl, ***P=0.001 for F1M1-Fc+ versus Ctrl, P=0.055 for F1M1-Fc+ versus F1M1-Fc-, P=0.416 for F1M1-Fc+ versus F1M1, P=0.066 for F1M1 versus F1M1-Fc- (mixed-effects multiple linear regression test). (B) Mean tumor volume at day 48. n=7 for Ctrl (rituximab); n=10 for F1M1; n=8 for F1M1-Fc-; n=9 for F1M1-Fc+. ***P=0.0003 for all groups (Kruskal-Wallis), *P=0.0289 for F1M1-Fc- versus Ctrl, ***P=0.0007 for F1M1 versus Ctrl, ***P=0.0002 for F1M1-Fc+ versus Ctrl, **P=0.0041 for F1M1-Fc+ versus F1M1-Fc-, P=0.0947 for F1M1-Fc+ versus F1M1, P=0.2114 for F1M1 versus F1M1-Fc- (Mann-Whitney t-test); data are the mean ± SEM. (C) Kaplan-Meier survival analysis. n=7 for Ctrl (rituximab); n=10 for F1M1; n=8 for F1M1-Fc-; n=9 for F1M1-Fc+. *P=0.0275 for F1M1-Fc- versus Ctrl, **P=0.0011 for F1M1 versus Ctrl, ***P=0.0003 for F1M1-Fc+ versus Ctrl, *P=0.0253 for F1M1-Fc+ versus F1M1- Fc-, P=0.3093 for F1M1 versus F1M1-Fc-, P=0.0953 for F1M1-Fc+ versus F1M1 (Log-rank Mantel-Cox test). (D) Mouse weight monitoring. n=7 for Ctrl (rituximab); n=10 for F1M1; n=8 for F1M1-Fc-; n=9 for F1M1-Fc+. Data are the mean ± SEM. Figure 10. F1M1-Fc+ triggers recruitment and activation of NK cells to induce ADCC in MDA-MB-231 cell xenografts. (A) Tumor growth. MDA-MB-231 cells were subcutaneously injected in nude mice. When tumor volume reached 50 mm3, mice were treated with F1M1 (n=11), F1M1-Fc- (n=10), F1M1-Fc+ (n=11), or rituximab (15mg/kg) (Ctrl; n=9), three times per week for 30 days. At day 48, mice were sacrificed. Tumor volume (in mm3) is shown as the mean ± SEM. **P=0.003 for F1M1-Fc+ versus Ctrl, P=0.066 for F1M1 versus Ctrl, P=0.353 for F1M1-Fc- versus Ctrl, P=0.061 for F1M1-Fc+ versus F1M1-Fc-, P=0.413 for F1M1 versus F1M1-Fc-, P=0.995 for F1M1-Fc+ versus F1M1 (mixed-effects multiple linear regression test). (B) Mean tumor volume at day 48. n=9 for Ctrl (rituximab); n=11 for F1M1; n=10 for F1M1-Fc-; n=11 for F1M1-Fc+. **P=0.0044 for all groups (Kruskal-Wallis), Mean ± SEM. *P=0.0368 for F1M1- Fc- versus Ctrl, *P=0.020 for F1M1 versus Ctrl, ***P=0.0005 for F1M1-Fc+ versus Ctrl, *P=0.0368 for F1M1-Fc+ versus F1M1-Fc-, P=0.6047 for F1M1 versus F1M1-Fc-, P=0.2234 for F1M1 versus F1M1-Fc+ (Mann-Whitney t-test). (C) NK cell recruitment at day 48. The percentage of NKp46+ NK cells was quantified by FACS and expressed relative to all living cells (n=7 for F1M1-Fc-; n=7 for F1M1; n=9 for F1M1-Fc+; n=8 for Ctrl). **P=0.0075 for all groups (Kruskal-Wallis), **P= 0.0053 for F1M1-Fc+ versus Ctrl, **P=0.0033 for F1M1-Fc+ versus F1M1-Fc-, *P=0.0295 for F1M1-Fc+ versus F1M1, P=0.4359 for F1M1-Fc- versus F1M1 (Mann-Whitney t-test). (D) Quantification of Eomes mRNA expression at day 48. Total RNA was extracted from MDA-MB-231 tumor cell xenografts at treatment end, and Eomes expression level was analyzed by RT-qPCR. Data are the mean ± SEM relative to Rps9 expression (n=8 for F1M1-Fc-; n=8 for F1M1; n=8 for F1M1-Fc+; n=7 for Ctrl). **P=0.0016 for all groups (Kruskal-Wallis), ***P=0.0006 for F1M1-Fc+ versus Ctrl, ***P=0.0006 for F1M1-Fc+ versus F1M1-Fc-, *P=0.0281 for F1M1 versus F1M1-Fc+, P=0.0650 for F1M1 versus F1M1-Fc- (Mann-Whitney t-test). (E) NK cell degranulation at day 48. The percentage of CD107a+ degranulating NK cells was quantified by FACS and expressed relative to all NKp46+ NK cells (n=7 for F1M1-Fc-; n=7 for F1M1; n=9 for F1M1-Fc+; n=7 for Ctrl). *P=0.0107 for all groups (Kruskal-Wallis), *P= 0.0464 for F1M1-Fc+ versus Ctrl, **P=0.0079 for F1M1-Fc+ versus F1M1-Fc-, **P=0.0021 for F1M1-Fc+ versus F1M1, P=0.3176 for F1M1 versus F1M1-Fc- (Mann-Whitney t-test). (F) Granzyme B+ NK cells at day 48. The percentage of granzyme-positive (GZMB+) activated NK cells was quantified by FACS and expressed relative to all NKp46+ NK cells (n=7 for F1M1-Fc-; n=7 for F1M1; n=9 for F1M1-Fc+; n=7 for Ctrl). *P=0.0119 for all groups (Kruskal-Wallis), **P= 0.0079 for F1M1-Fc+ versus Ctrl, *P=0.0289 for F1M1 versus Ctrl, *P=0.0418 for F1M1-Fc+ versus F1M1-Fc-, *P=0.0262 F1M1 versus F1M1-Fc-, P>0.9999 for F1M1-Fc+ versus F1M1 (Mann-Whitney t-test). G Figure 11: Cath-D expression in MDA-MB-231 cell xenografts of mice treated with F1M1-Fc-, F1M1, or F1M1-Fc+ (A) Representative images of MDA-MB-231 cell xenograft sections showing cath- D expression. Cath-D was monitored in MDA-MB-231 cell xenografts from mice treated with F1M1-Fc-, F1M1, or F1M1-Fc+ (from figure 10B) (n=9 for F1M1-Fc-; n=8 for F1M1; n=5 for F1M1-Fc+; n=9 for Ctrl) by IHC using an anti-cath-D (clone CTD19) antibody. Scale bar, 50 µm. (B) Quantification of cath-D expression in MDA-MB-231 cell xenograft sections. (n=9 for F1M1-Fc-; n=8 for F1M1; n=5 for F1M1-Fc+; n=9 for Ctrl). P=0.8815, Kruskal-Wallis test. (C) Cath-D expression in MDA-MB-231 cell xenografts. Cath-D expression was determined in MDA-MB-231 cell xenografts from mice treated with F1M1-Fc-, F1M1, or F1M1-Fc+ (from figure 4B) (n=3 for F1M1-Fc-; n=3 for F1M1; n=3 for F1M1-Fc+; n=3 for Ctrl). Whole cytosols (10 µg proteins) were immunoblotted with the mouse monoclonal (#610801) (to detect mature cath-D) and rabbit polyclonal (H-75) (to detect pro-cath-D) anti-cath-D antibodies. HSC70 (clone B-6) was used as loading control. Mr, relative molecular mass (kDa). Figure 12. NK cell depletion impairs F1M1-Fc+ therapeutic efficacy in MDA-MB- 231 cell xenografts. (A) Treatment schedule for NK cell depletion. MDA-MB-231 cells were subcutaneously injected in nude mice. At day 9 after MDA-MB-231 injection, half of the mice started the treatment with anti-asialo GM1 antibodies (αGM1, 50 µl by ip injection twice per week) to deplete NK cells. When tumor volume reached 50 mm3 (day 15), mice were treated with F1M1-Fc+ (n=8), or control rituximab (Ctrl; n=8) (15mg/kg), three times per week for 30 days, in the absence or presence of αGM1. At day 48, mice were sacrificed. (B) Tumor Growth. Tumor growth (in mm3) is shown as the mean ± SEM. (n=8 for F1M1-Fc+; n=8 for F1M-Fc+ + αGM1; n=8 for Ctrl; n=8 for Ctrl + αGM1). P=0.449 for Ctrl + αGM1 versus Ctrl, ***P<0.001 for F1M1-Fc+ versus Ctrl, ***P<0.001 for F1M1-Fc+ versus Ctrl + αGM1, *P=0.015 for F1M1-Fc+ + αGM1 versus F1M1-Fc+, *P=0.021 for F1M1-Fc+ + αGM1 versus Ctrl + αGM1, P=0.127 for F1M1-Fc+ + αGM1 versus Ctrl (mixed-effects multiple linear regression test). (C) Mean tumor volume at day 48. Mean ± SEM. (n=8 for F1M1-Fc+; n=8 for F1M-Fc+ + αGM1; n=8 for Ctrl; n=8 for Ctrl + αGM1). **P=0.0018 for all groups (Kruskal- Wallis), ***P=0.0003 for F1M1-Fc+ versus Ctrl, P=0.8785 for Ctrl + αGM1 versus Ctrl, **P=0.0019 for F1M1-Fc+ versus Ctrl + αGM1, *P=0.0281 for F1M1-Fc+ + αGM1 versus F1M1-Fc+, P=0.0881 for F1M1-Fc+ + αGM1 versus Ctrl, P=0.1605 for F1M1-Fc+ + αGM1 versus Ctrl + αGM1 (Mann-Whitney t-test). (D) NK cell recruitment at day 48. The number of NKp46+ NK cells in tumors was quantified by FACS and normalized per mg of tumor with precision count beads (n=8 for F1M1-Fc+; n=8 for F1M-Fc+ + αGM1; n=7 for Ctrl; n=8 for Ctrl + αGM1). ****P<0.0001 for all groups (Kruskal-Wallis), *P=0.0312 for F1M1-Fc+ versus Ctrl, ***P=0.0006 for Ctrl + αGM1 versus Ctrl, ***P=0.0002 for F1M1-Fc+ versus Ctrl + αGM1, ***P=0.0002 for F1M1-Fc+ + αGM1 versus F1M1-Fc+, ***P=0.0008 for F1M1-Fc+ + αGM1 versus Ctrl, P=0.1621 for F1M1-Fc+ + αGM1 versus Ctrl + αGM1 (Mann-Whitney t-test). Figure 13: Analysis of neutrophils and blood counts in MDA-MB-231 cell xenografted nude mice treated with F1M1-Fc+. (A) Neutrophils in peripheral blood. The percentage of neutrophils in peripheral blood samples of nude mice treated with F1M1-Fc+ or rituximab (control, Ctrl) was quantified by FACS at day 45 (n=7 for F1M1-Fc+; n=8 for Ctrl). Neutrophils were defined as CD45+ CD3- Ly6G+ cells. (B) Blood counts. Blood samples from mice treated with F1M1-Fc+ or rituximab (Ctrl) were analyzed using the scil Vet abc Plus+ system at day 48 (scil Animal Care Co) (n=8 for F1M1-Fc+; n=8 for Ctrl). White blood cells (top left panel), platelets (top right panel), red blood cells (bottom left panel) and hemoglobin (bottom right panel) were quantified.Figure 14. The lead F1M1-Fc+ anti-Cath-D antibody inhibits growth of TNBC-PDXs and improves mouse survival. (A) Therapeutic effects of F1M1-Fc+ in mice xenografted with PDX B1995. Mice were xenografted with PDX B1995 and when tumor volume reached 100 mm3 (day 28), mice were treated with F1M1-Fc+ (15 mg/kg) or rituximab (Ctrl) (15 mg/kg) three times per week. Mice were sacrificed when tumor volume reached 1500 mm3, and the corresponding tumor growth curves were stopped (left panel). Tumor volume (in mm3) is shown as the mean ± SEM; n=12 for Ctrl (rituximab); n=12 for F1M1-Fc+. ***P<0.001 for F1M1-Fc+ (mixed-effects multiple linear regression test). Mean tumor volume at day 48 (middle panel). Mean ± SEM; n=12 for Ctrl (rituximab); n=12 for F1M1-Fc+. ***P=0.0001 for F1M1-Fc+ versus Ctrl (Mann- Whitney test). Kaplan-Meier survival analysis (right panel). n=12 for Ctrl (rituximab); n=12 for F1M1-Fc+. ***P=0.0002 for F1M1-Fc+ versus Ctrl (Log-rank Mantel-Cox test). (B) Therapeutic effects of F1M1-Fc+ in mice xenografted with PDX B3977. Mice were xenografted with PDX B3977 and when tumor volumes reached 100 mm3 (day 25), mice were treated with F1M1-Fc+ (15 mg/kg) or rituximab (Ctrl) (15 mg/kg) three times per week. Mice were sacrificed when tumor volume reached 1500 mm3, and the corresponding tumor growth curves were stopped (left panel). Tumor volume (in mm3) is shown as the mean ± SEM; n=11 for Ctrl (rituximab); n=11 for F1M1-Fc+. ***P <0.001 for F1M1-Fc+ (mixed-effects multiple linear regression test). Mean tumor volume at day 42 (middle panel). Mean ± SEM; n=11 for Ctrl (rituximab); n=11 for F1M1-Fc+. **P=0.0018 for F1M1-Fc+ versus Ctrl (Mann-Whitney test). Kaplan-Meier survival analysis (right panel). n=11 for Ctrl (rituximab); n=11 for F1M1- Fc+. **P=0.0024 for F1M1-Fc+ versus Ctrl (Log-rank Mantel-Cox test). Figure 15. Therapeutic efficacy of the anti-Cath-D F1M1-Fc+ antibody in combination with paclitaxel in mice xenografted with MDA-MB-231 cells. (A) Tumor growth in mice treated with F1M1-Fc+ and/or paclitaxel (1 mg/kg; PTX LD). MDA-MB-231 cells were subcutaneously injected in nude mice. When tumor volume reached 50 mm3 (day 15 post-graft), mice were treated with F1M1-Fc+ (15mg/kg; three times per week) (n=9), PTX LD (1 mg/kg; once per week) (n=8), F1M1-Fc+ (15mg/kg; three times per week) + PTX LD (1 mg/kg; once per week) (n=9), or rituximab (three times per week) + saline (once per week; all ip) (Ctrl; n=7) for 37 days. Mice were sacrificed when tumor volume reached 2000 mm3. Tumor volume (in mm3) is shown as the mean ± SEM. *P=0.039 for F1M1- Fc+ versus Ctrl, P=0.689 for PTX LD versus Ctrl, *P=0.032 for PTX LD+F1M1-Fc+ versus Ctrl, P=0.092 for PTX LD+F1M1-Fc+ versus PTX LD, P=0.88 for PTX LD+F1M1-Fc+ versus F1M1-Fc+ (mixed-effects multiple linear regression test). PTX LD; low dose of paclitaxel (1 mg/kg). (B) Mean tumor volume at day 52 in mice receiving F1M1-Fc+ and/or paclitaxel (1 mg/kg; PTX LD). *P=0.0189 for all groups (Kruskal-Wallis), P=0.0712 for F1M1-Fc+ versus Ctrl; P=0.3211 for PTX LD versus Ctrl; **P=0.0020 for PTX LD+F1M1-Fc+ versus Ctrl; *P=0.0293 for PTX LD+F1M1-Fc+ versus PTX LD; P=0.5302 for PTX LD+F1M1-Fc+ versus F1M1-Fc+ (Mann-Whitney test); Data are the mean ± SEM. (C) Kaplan-Meier survival analysis in mice receiving F1M1-Fc+ and/or paclitaxel (1 mg/kg; PTX LD). *P=0.0313 for F1M1-Fc+ versus Ctrl, P=0.0875 for PTX LD versus Ctrl, ***P=0.0009 for PTX LD+F1M1- Fc+ versus Ctrl, *P=0.0216 for PTX LD+F1M1-Fc+ versus PTX LD, P=0.3714 for PTX LD+F1M1-Fc+ versus F1M1-Fc+ (Log-rank Mantel-Cox test). (D) Tumor growth in mice receiving F1M1-Fc+ and/or paclitaxel (4 mg/kg; PTX MD). MDA-MB-231 cells were subcutaneously injected in nude mice. When tumor volume reached 50 mm3 (day 15 post-graft), mice were treated with F1M1-Fc+ (15mg/kg; three times per week) (n=9), PTX MD (4 mg/kg; once per week) (n=8), F1M1-Fc+ (15mg/kg; three times per week) + PTX MD (4 mg/kg; once per week) (n=7), or rituximab (three times per week) + saline (once per week) (Ctrl; n=7) for 37 days. Mice were sacrificed when tumor volume reached 2000 mm3. Tumor volume (in mm3) is shown as the mean ± SEM. *P=0.039 for F1M1-Fc+ versus Ctrl, *P=0.042 for PTX MD versus Ctrl, *P=0.013 for PTX MD+F1M1-Fc+ versus Ctrl, P=0.107 for PTX MD+F1M1-Fc+ versus PTX MD, P=0.857 for PTX MD+F1M1-Fc+ versus F1M1-Fc+ (mixed-effects multiple linear regression test). PTX MD; medium dose of paclitaxel (4 mg/kg). (E) Mean tumor volume at day 52 in mice receiving F1M1-Fc+ and/or paclitaxel (4 mg/kg; PTX MD). *P=0.0200 for all groups (Kruskal-Wallis), P=0.0712 for F1M1-Fc+ versus Ctrl; *P=0.0289 for PTX MD versus Ctrl; **P=0.0012 for PTX MD+F1M1-Fc+ versus Ctrl; P=0.1520 for PTX MD+F1M1-Fc+ versus PTX MD; P=0.4698 for PTX MD+F1M1-Fc+ versus F1M1-Fc+ (Mann- Whitney test); Data are the mean ± SEM. (F) Kaplan-Meier survival analysis in mice receiving F1M1-Fc+ and/or paclitaxel (4 mg/kg; PTX MD). *P=0.0313 for F1M1-Fc+ versus Ctrl, *P=0.0312 for PTX MD versus Ctrl, ***P=0.001 for PTX MD+F1M1-Fc+ versus Ctrl, *P=0.0410 for PTX MD+F1M1-Fc+ versus PTX MD, P=0.2010 for PTX MD+F1M1-Fc+ versus F1M1-Fc+ (Log-rank Mantel-Cox test). Figure 16. Therapeutic efficacy of the anti-Cath-D F1M1-Fc+ antibody alone and in combination with enzalutamide in mice xenografted with SUM159 cells. (A) Tumor growth in mice receiving F1M1-Fc+ monotherapy. SUM159 cells were subcutaneously injected in nude mice. When tumor volume reached 50 mm3, mice were treated with F1M1-Fc+ (n=9) (15mg/kg), or rituximab (Ctrl; n=9) (15mg/kg), three times per week for 30 days. Mice were sacrificed when tumor volume reached 1000 mm3. Tumor volume (in mm3) is the mean ± SEM. ***P <0.001 (mixed-effects multiple linear regression test). (B) Mean tumor volume at day 48 in mice receiving F1M1-Fc+ monotherapy. Mean ± SEM. ****P <0.0001 for F1M1-Fc+ versus Ctrl (Mann-Whitney test). (C) Kaplan-Meier survival analysis in mice receiving F1M1-Fc+ monotherapy. **P=0.0024 for F1M1-Fc+ versus Ctrl (Log-rank Mantel-Cox test). (D) Weight monitoring in mice receiving F1M1-Fc+ monotherapy. Mean ± SEM. Rituximab (Ctrl; n=9), F1M1-Fc+ (n=9). (E) Tumor growth in mice receiving the F1M1-Fc++enzalutamide combination therapy. SUM159 cells were subcutaneously injected in nude mice. When tumor volume reached 50 mm3, mice were treated with rituximab + corn oil (Ctrl; n=8), F1M1-Fc+ (n=8) (15mg/kg, twice per week), enzalutamide (Enza; 30mg/kg, five times per week, per os) (n=8), or F1M1-Fc+ (15mg/kg, twice per week, ip) + enzalutamide (Enza; 30mg/kg, five times per week, per os) (n=8) for 32 days. Mice were sacrificed when tumor volume reached 1500 mm3. Tumor volume (in mm3) is shown as the mean ± SEM. ***P=0.001 for F1M1-Fc+ versus Ctrl, P=0.5 for Enza versus Ctrl, *P=0.019 for Enza versus F1M1-Fc+, **P=0.003 for Enza+F1M1-Fc+ versus Ctrl, **P=0.008 for Enza+F1M1-Fc+ versus Enza, P=0.887 for Enza+F1M1-Fc+ versus F1M1-Fc+ (mixed-effects multiple linear regression test). (F) Mean tumor volume at day 50 in mice receiving the F1M1-Fc+ + enzalutamide combination. Mean ± SEM. ***P=0.0005 for all groups (Kruskal-Wallis), **P=0.0019 for F1M1-Fc+ versus Ctrl, *P=0.02 for Enza versus Ctrl, ***P=0.0002 for Enza+F1M1-Fc+ versus Ctrl, ** P=0.0092 for Enza+F1M1-Fc+ versus Enza, P=0.0985 for Enza+F1M1-Fc+ versus F1M1-Fc+ (Mann-Whitney test). (G) Kaplan-Meier survival analysis in mice receiving the F1M1-Fc+ + enzalutamide combination. *P=0.0126 for F1M1-Fc+ versus Ctrl, *P=0.0196 for Enza versus Ctrl, ****P <0.0001 for Enza+F1M1-Fc+ versus Ctrl, *P=0.017 for Enza+F1M1-Fc+ versus Enza, ** P=0.0091 for Enza+F1M1-Fc+ versus F1M1-Fc+ (Log-rank Mantel-Cox test). (H) Weight monitoring in mice receiving the F1M1-Fc+ + enzalutamide combination. Mean ± SEM. Rituximab (Ctrl; n=8), F1M1-Fc+ (n=8); Enza (n=8), Enza+F1M1-Fc+ (n=8). Figure 17: F1M1-ADCs inhibit tumor growth of 4-hydroxytamoxifen-resistant LCC2-MCF7 breast cancer cells and improve mouse survival (A) Effect of F1M1-MMAF and RTX-MMAF on tumor growth. F1M1 anti-cath-D or rituximab (RTX, control) were bioconjugated, via some of the eight cysteines forming interchain disulfide bridges, to auristatin F (MMAF) through a non-cleavable linker with a drug- to-antibody ratio (DAR) of 3.6 (F1M1-MMAF) and 3.4 (RTX-MMAF), according to patent WO2015004400. MCF7-LCC2 cells (5106 cells), a 4-hydroxytamoxifen resistant human breast cancer cell line, were subcutaneously injected in nude mice. When tumor volume reached 100 mm3, mice were treated intravenously three times with F1M1-MMAF (n=10), or RTX-MMAF (n=10) at days 17 (10mg/kg), 24 (5mg/kg) and 31 (5mg/kg) post-graft. Mice were sacrificed when tumor volume reached 1000 mm3. Tumor volume (in mm3) is shown as the mean ± SEM. **P=0.004 for F1M1-MMAF versus RTX-MMAF (mixed-effects multiple linear regression test). (B) Mean tumor volume at day 64 for F1M1-MMAF and RTX-MMAF. n=10 for F1M1-MMAF; n=10 for RTX-MMAF; ****P<0.0001 for F1M1-MMAF versus RTX-MMAF (Student t-test); data are the mean ± SEM. (C) Kaplan-Meier survival analysis for F1M1- MMAF and RTX-MMAF. n=10 for F1M1-MMAF; n=10 for RTX-MMAF; ****P<0.0001 for F1M1-MMAF versus RTX-MMAF (Log-rank Mantel-Cox test). (D) Effect of F1M1- MMAE and RTX-MMAE on tumor growth. F1M1 anti-cath-D or rituximab (control, RTX) were bioconjugated, via some of the eight cysteines forming interchain disulfide bridges, to auristatin E (MMAE) through a valine-citrulline cleavable linker with a drug-to-antibody ratio (DAR) of 3.8 (F1M1-MMAE) and 3.5 (RTX-MMAE), according to patent WO2015004400. MCF7-LCC2 cells (5106 cells) were subcutaneously injected in nude mice. When tumor volume reached 100 mm3, mice were treated intravenously three times with F1M1-MMAE (n=10), or RTX-MMAE (n=10) at day 17 (10mg/kg), 24 (5mg/kg) and 31 (5mg/kg) post-graft. Mice were sacrificed when tumor volume reached 1000 mm3. Tumor volume (in mm3) is shown as the mean ± SEM. *P=0.014 for F1M1-MMAE versus RTX-MMAE (mixed-effects multiple linear regression test). (E) Mean tumor volume at day 64 for F1M1-MMAE and RTX- MMAE. n=10 for F1M1-MMAE; n=10 for RTX-MMAE; **P=0.0053 for F1M1-MMAE versus RTX-MMAE (Student t-test); data are the mean ± SEM. (F) Kaplan-Meier survival analysis for F1M1-MMAE and RTX-MMAE. n=10 for F1M1-MMAE; n=10 for RTX- MMAE; *****P<0.0001 for F1M1-MMAE versus RTX-MMAE (Log-rank Mantel-Cox test). (G) Mouse weight monitoring for F1M1-MMAF, RTX-MMAF, F1M1-MMAE and RTX- MMAE. n=10 for F1M1-MMAF, n=10 for RTX-MMAF, n=10 for F1M1-MMAE and n=10 for RTX-MMAE. Data are the mean ± SEM. EXAMPLES: As used in EXAMPLES, the term "F1 antibody" refers to an antibody directed and having specificity for Cath-D previously generated by the inventors and described in Table 2, WO2016/188911 and Ashraf et al.2019 (22). F1 antibody was also disclosed in the example of the patent WO2020/127411 (an typing error was made in the definition of CDR3 sequence between GDS and YDS in WO2020/127411). ScFv F1 Sequence (defined by IMGT unique numbering for IgG) Domains VH EVQLVESGGSLVKPGGSLRLSCAASGFTSNNYMNWVRQAPGKG LEWISYISGSSRYISYADFVKGRFTISRDNATNSLYLQMNSLRAED TAVYYCVRSSNSGGMDVWGRGTLVTVSS (SEQ ID NO: 8) H-CDR1 GFTFSNNY (SEQ ID NO: 2) H-CDR2 ISGSSRYI (SEQ ID NO: 3) H-CDR3 VRSSNSGGMDV (SEQ ID NO: 4) VL QSVLTQPASVSGSPGQSITISCAGTSSDVGGYYGVSWYQQHPGKA PKLMIYYDSYRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYC SSYTSNSTRVFGGGTKLAVL (SEQ ID NO: 9) L-CDR1 SSDVGGYYG (SEQ ID NO: 6) L-CDR2 YDS L-CDR3 SSYTSNSTRV (SEQ ID NO: 10) Table 2: Sequences of ScFv F1 antibody. The term “F1M1” refers to the antibody of the present invention comprising a heavy chain wherein the variable domain has a sequence set forth as SEQ ID NO:1 and a light chain has a heavy chain wherein the variable domain has a sequence set forth as SEQ ID NO:5. EXAMPLE 1: Anti-Cathepsin D antibody F1M1 triggers both innate and adaptive anti-tumor immunity in breast cancer Material & Methods Reagents The anti-mouse Cath-D antibody against the 52-, 48-, 34-kDa forms was from Bio- Techne (#AF1029, R&D Systems - Minneapolis, MN). The HRP-conjugated anti-mouse F(ab)2 (#115-036-072) was from Jackson ImmunoResearch (West Grove, PA). The substrate reagent for HRP was purchased from Bio-Techne (R&D Systems, #DY999). The anti-HSC70 antibody (#sc-7293) was from Santa Cruz BioTechnology (Dallas, TX). The fluorescent-conjugated anti- mouse (#922-32210) and anti-goat (#926-68071) antibodies were from LI-COR Biosciences (Lincoln, NE). Injectable D-luciferin (#122799-5) was from Perkin Elmer (Waltham, MA). The mouse IgG2a isotype control antibody (C1.18.4, #BE0085) was from BioXCell (Lebanon, NH). Collagenase IV (#C5138) and DNase I (#11284932001) were from Sigma-Aldrich (St Louis, MO). Mouse Fc Block (#130-097-575) was from Miltenyi Biotec (Bergisch Gladbach, Germany). Hoechst (#33342) was from Thermo Fisher Scientific (Waltham, MA). The AF488- conjugated anti-human IgG (#A11013) was from Life Technologies (Carlsbad, CA). The fluorescent-conjugated antibodies against CD11b (clone M1/70, #101205), CD3 (clone 17A2, #100205), PD-L1 (clone 10F.9G2, #135228), F4/80 (clone QA17A29, #157308), CD16 (clone S17014E, #158005), Ly6G (clone 1A8, #127621), NKp46 (clone 29A1.4, #1377631), CD11c (clone N418, #117330), PD-1 (clone 29F.1A12, #103044), CD206 (clone C068C2, #141721), B220/CD45R (clone RA3-632, #103241), and LAG3 (clone C9B7W, #125219) were from Biolegend (San Diego, CA). The antibody against Ly6C (clone REA796, #130-111-920) was from Miltenyi Biotec; the antibodies against CD4 (clone GK1.5, #565974), CD8 (clone 53-6.7, #748535), and CD45 (clone 30-F11, #612975) were from BD Biosciences (Franklin Lakes, NJ). Viakrome IR 808 (#C36628) was from Beckman Coulter (Brea, CA). Mouse F1M1 IgG2a, mouse F1 IgG2a and human F1M1 IgG1 were constructed from the scFv (F1, F1M1) sequences using gene synthesis, expressed in the Chinese hamster ovary cell line and purified on protein- A HiTrap columns (GE Healthcare) by Evitria AG (Schlieren, Switzerland). Cell lines The E0771Luc cell line transformed to constitutively express luciferase as reporter, was kindly provided by Dr. C-L. Tomasetto (IGBMC, Strasbourg, France). The coding sequence of the luciferase reporter gene luc2 (Photinus pyralis) was PCR-amplified from the pGL4.50[luc2/CMV/Hygro] vector (#E1310, Promega, Madison, WI) and the flanking XhoI restriction sites were added. The digested PCR fragment was sub-cloned into the SalI site of pLENTI PGK Blast DEST Vector (Plasmid #19065, Addgene, Cambridge, MA). To generate lentiviral particles, the pLENTI PGK Blast vector was co-transfected with three packaging plasmids (pLP1, pLP2, and pLP/VSVG) (Invitrogen, Carlsbad, CA) in the 293T cell line. Then, E0771 cells were incubated with viral particles, supplemented with 10 μg/ml polybrene and 20 mM HEPES. Luciferase expression was assessed in blasticidin (5 µg/ml)-resistant cells by bioluminescence. E0771Luc cells were cultured in RPMI1640 with 10% fetal calf serum (FCS) (Eurobio Scientific – Les Ulis, France) and 10 mM HEPES, pH=7.5 (Gibco). The 4T1 cell line, kindly provided by A. Pèlegrin (IRCM, Montpellier, France), was cultured in DMEM/GlutaMAX, 10% FCS (Eurobio Scientific). All cell lines were mycoplasma-free, determined using the MycoAlertTM detection kit (Lonza, Switzerland). To produce 48h- supernatant containing secreted Cath-D, cells were grown to 60% confluence, and left in medium with FCS for 48h. Conditioned medium was centrifuged at 800 x g for 5min. Sandwich ELISA First, 96-well plates were coated with anti-Cath-D antibody (#AF1029, Bio-Techne) in PBS (100 ng/well) at 4°C overnight. After blocking non-specific sites with PBS/0.1% Tween 20/1% BSA, Cath-D-containing supernatants from E0771Luc were added at 37°C for 2h. After washes in PBS/0.1% Tween 20, two-fold serial dilutions of F1 or F1M1 (initial concentration: 4224 nM) in PBS were added at 37°C for 2h, followed by washes in PBS/0.1% Tween 20, and incubation with a HRP-conjugated anti-mouse F(ab)2 (1/2000) at 37°C for 2h. Then, plates were rinsed four times in PBS/0.1% Tween 20, and 50 µl/well of TetraMethylBenzidine substrate (#34028, ThermoFisher) was added at room temperature in the dark for 10-30min. The reaction was stopped with 50 µl/well of 1M hydrochloric acid and optical density was read at a wavelength of 450 nm. GraphPad Prism 8 (GraphPad Software, RRID:SCR_002798), was used to calculate the EC50 of the antibodies. Five independent assays with three replicates were performed for cancer cell lines. Western blotting Cell lysates were harvested in lysis buffer (50 mM HEPES pH 7.5, 150 mM NaCl, 10% glycerol, 1% Triton X-100; 1.5 mM MgCl2, 1 mM EGTA) supplemented with cOMPLETE™ protease inhibitor cocktail (#4693116001, Roche, Switzerland) at 4°C for 10min. Protein concentration was determined with the DC protein assay (Bio-Rad). Then, protein samples (20 µg of cell lysates, 40 µL of cell supernatants, 5 µg of tumor lysates) were separated on 13.5% SDS-PAGE and transferred to PVDF membranes (#IPFL85R, Merck Millipore, Burlington, MA) by liquid-phase transfer at 4°C for 90min. After transfer, membranes were blocked with 100 mM Tris buffer, pH 7.4, 150 mM NaCl, 0.1% Tween 20 (TBT-T) and 3% BSA at room temperature for 1h, followed by incubation with anti-HSC70 or anti-Cath-D antibodies at 4°C overnight. After TBT-T washes, membranes were incubated with secondary fluorescent- conjugated anti-mouse or anti-goat antibodies in TBT-T/3% BSA at room temperature for 1h, followed by image acquisition with a LI-COR Odyssey imager, according to the manufacturer’s instructions. In vivo studies Animals Mouse experiments were performed in compliance with the French regulations and ethical guidelines for experimental animal studies in an accredited establishment (Agreement No. #31135-2021042212479661). Animal studies are reported in compliance with the ARRIVE guidelines (Percie du Sert et al., 2020), and with the recommendations made by the British Journal of Pharmacology (Lilley et al., 2020). Female C57BL/6N and BALB/c (6-weeks old) were purchased from Charles River (Wilmington, MA). Mice used in the experiments were 8- weeks old (20g). Mice (5 mice per cage) were kept in individually ventilated cages (transparent and with top filter-isolator) with standard bedding at a constant temperature of 23 °C and 40- 60% humidity with 12h light/darkness cycle in a specific pathogen-free conditions with food and water at will. All the animal studies were designed to generate groups of equal size using randomisation and blinded analysis. The different group sizes are due to the purpose of unexpected individual losses during the process. Orthotopic and subcutaneous tumour growth E0771Luc cells (2.5× 105 in PBS) were injected orthotopically in the mammary fat pad between the fourth and fifth mammary glands of 8-week-old female C57BL/6N mice (Charles River, Wilmington, MA). After 2 days, tumor-bearing mice with similar E0771Luc cell bioluminescence were randomized in three treatment groups: isotype control C1.18.4 (15 mg/kg), F1 (15 mg/kg), or F1M1 (15 mg/kg) (all by intraperitoneal injection three times per week until day 33). For bioluminescence harmonization, at day 2 post-graft, 200 µL luciferin (10mg/mL) was injected in anaesthetized grafted C57BL/6N mice followed by imaging using an IVIS Lumina II camera (Caliper LifeSciences, Waltham, MA). Bioluminescence was quantified with the Living Image® v4.5.2.18424 software. Tumors were measured using a caliper and volume was calculated with the formula: ^^^^ =( ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ℎ × ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ℎ × ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ ^^^^ℎ 2 . Mice were sacrificed and tumors, draining lymph nodes (dLNs), and peripheral blood lymphocytes (PBLs) were collected for immunophenotyping. Tumor samples were frozen in liquid nitrogen for RNA extraction. Quantitative RT-PCR Total RNA was extracted using the Quick-RNA Mini Prep kit (#R1054, Zymo Research – Irvine, CA), according to the manufacturer’s instructions. Reverse transcription of total RNA was performed at 37°C using the Transcriptase Inverse SuperScript III (Invitrogen) and random hexanucleotide primers (Promega). Real-time quantitative PCR analyses were performed on a Light Cycler 480 ONEGreen FAST pPCR Premix (#OZYA008-200XL, Ozyme – Saint-Cyr- l’Ecole, France) and a LightCycler 480 apparatus (Roche Diagnostics – Bâle, Switzerland). The PCR product integrity was checked by melting curve analysis. Expression data were normalized to the amplification data for the reference gene RPS9. The experiments were performed in triplicate with more than 5 animal per cohort. Isolation of tumor-infiltrating cells and immunophenotyping by 17-color flow cytometry Tumors were enzymatically and mechanically digested in gentleMACS™ C-Tubes (#130-093-237, Miltenyi Biotec) that contained a mixture of collagenase IV (1 mg/ml) and DNase I (200 U/ml) in RPMI using a gentleMACS™ Octo Dissociator (Miltenyi Biotec) at 37°C (two incubations of 19.5 min and 5.5 min). After digestion, tumor suspensions were passed through a 70 μm nylon cell strainer (#22-363-548, Fisher Scientific, Waltham, MA), centrifuged and resuspended in FACS buffer (PBS pH 7.2, 0.5% FBS, and 0.02% sodium azide). dLNs were mechanically dissociated in PBS, and re-suspended in FACS buffer. To obtain PBLs, red blood cells were removed by adding 10 volumes of ammonium-chloride- potassium lysing buffer, and PBLs were recovered by centrifugation, washed with PBS and resuspended in FACS buffer. Cells were blocked with FACS buffer containing 1% (v/v) of mouse Fc Block, and stained with fluorescent-conjugated antibodies, followed by washes in FACS buffer, and fixation with 1% paraformaldehyde in PBS. Samples were analyzed by flow cytometry using a Beckman and Coulter CytoFLEX LX 17-color flow cytometer. Immune cell populations from tumors were analyzed using the following 17-color flow cytometry panel: Viakrome IR 808, and antibodies against CD45, CD11b, F4/80, CD206, CD3, CD4, CD8, Ly6G, Ly6C, NKp46, CD11c, B220, CD86, CD16, PD1, and PDL1. Living immune cells were defined as Viakrome IR 808- and CD45+. TAMs were defined as CD45+ CD11b+ F4/80+; the M1-TAM subset as F4/80+ CD206- CD11c+; and the M2-TAM subset as F4/80+ CD206+. Conventional DCs (cDCs) were defined as CD45+ CD3- Ly6G- B220+ CD11c+/hi; the cDC2 subset as CD11c+/hi CD8- CD11b+; and the cDC1 subset as CD11c+/hi CD8+ CD11b-. B cells were defined as CD45+ F4/80- CD3- Ly6G- Ly6C- CD11c- B220+. NK cells were defined as CD45+ F4/80- CD3- Ly6C- Ly6G- CD11blo/+ NKp46+. T cells were defined as CD45+ F4/80- CD3+; CD8+ T cells as CD3+ CD8+; and CD4+ T cells as CD3+ CD4+. Functional markers (CD86, CD16, PDL1) were studied in the relevant immune cell subpopulations. Immune cells from PBLs and dLNs were analyzed using the following 17-color flow cytometry panel: Viakrome IR 808, and antibodies against CD45, CD3, CD4, CD8, CD11b, Ly6G, Ly6C, NKp46, CD11c, B220, CD16, PDL1, and LAG3. In PBLs, living immune cells were defined as Viakrome IR 808- and CD45+. T cells were defined as CD45+ CD3+; CD8+ T cells as CD3+ CD8+; and CD4+ T cells as CD3+ CD4+. NK cells were defined as CD45+ CD3- CD11b+/lo NKp46+. Functional markers (CD16, LAG3, PDL1) were studied in the relevant immune cell subpopulations. Events were analyzed with FlowJo 10.8.1. Fluorescence microscopy Paraffin-embedded E0771Luc cell graft tissue sections were deparaffined, rehydrated, rinsed and saturated in PBS with 5% FCS at 4°C overnight. Sections were incubated with 25 µg/mL F1M1 (human IgG1 format), followed by incubation with AF488-conjugated anti- human IgG (#A11013). Nuclei were stained with 0.5 µg/mL Hoechst 33342. Images were acquired with a x63 Plan-Apochromat objective and a Zeiss Axio Imager light microscope for each tumour tissue from 5 mice for E0771. Statistical analyses The data and statistical analysis comply with the recommendations of the British Journal of Pharmacology on experimental design and analysis in pharmacology (Curtis et al., 2018). Additional data from two different models were provided to validate the results. In our in vivo analysis, the size of independent groups was superior to 5 (exact numbers are provided in the figure legends). Results are expressed as mean ± SEM. A linear mixed regression model was used to determine the relationship between tumour growth and number of days post-graft. The variables included in the fixed part of the model were: number of days post-graft and treatment groups. Their interaction was also evaluated. Random intercepts and random slopes were included to take into account the time effect. The model coefficients were estimated by maximum likelihood. The statistical analyses for linear mixed regression model for tumour growth were done with STATA 13.0. The Mann-Whitney t test was used for pairwise comparisons. Statistical analysis were performed using GraphPad Prism 8 (GraphPad Software, RRID:SCR_002798). All outliers were included in data analysis. P <0.05 values were considered indicative of statistical significance. All studies followed the editorial on experimental design and analysis in pharmacology (Curtis et al., 2018) and followed the BJP checklist for Design and Analysis. Results Cath-D expression and secretion in preclinical mouse models of breast cancer Analysis of Cath-D expression and secretion showed expression mainly of the 48-kDa intermediate chain and to a lesser extent of the mature 34-kDa heavy chain in E0771 and 4T1 cells (TNBC) (data not shown). We observed the 52-kDa Cath-D precursor in the supernatant (SN) of E0771 (data not shown), but not of 4T1 cells (data not shown). Cath-D was also expressed in E0771 cell grafts (data not shown) where it showed intense and punctuate staining (data not shown) that mirrored the expression pattern in BC biopsy samples (12). These observations demonstrated that EO771Luc cell line is a suitable model to study the antitumor and immunomodulatory activity of anti-Cath-D antibodies in syngeneic mouse models of basal- like TNBC The mouse IgG2a-formatted F1 and Fab-aglycosylated F1M1 antibodies bind to secreted mouse Cath-D We formatted the anti-Cath-D IgG1 human antibody F1 (12) in the murine IgG2a format to investigate its anti-tumor effect and immunomodulatory activity in fully immunocompetent mouse models of BC. We also generated the new murine IgG2a anti-Cath-D antibody F1M1 with point mutations (one in VL CDR3 and one in VH FR3) to abrogate Fab N-glycosylation that may lead to the production of glycoforms associated with the risk of immunogenic responses in patients. The migration profiles of F1 and F1M1 showed that the molecular weight of the light and heavy chains was lower in F1M1 than F1 (data not shown). Sandwich ELISA using Cath-D secreted from E0771 cells showed that F1 and F1M1 had good binding capacities, with a better affinity for F1M1 (EC50=43.5 nM and 33.8 nM for F1 and EC50=0.9 nM and 0.2 nM for F1M1, respectively) (Fig. 1). The lower EC50 of F1M1 indicated that the two point mutations leading to Fab aglycosylation enhanced F1M1 capacity to recognize mouse Cath-D, possibly because VL-CDR3 glycosylation decreased F1 affinity due to steric hindrance. Therefore, both F1 and F1M1 were investigated in vivo. Immunological profile of the E0771 TNBC model To investigate the anti-tumoral and immunomodulatory impact of our anti-Cath-D antibodies, we established an orthotopic TNBC model by grafting basal-like p53-mutated E0771 cells in C57BL/6 mice, This Cath-D-secreting cell line readily formed tumors upon grafting (data not shown). We then analyzed tumor-infiltrating immune cells in untreated E0771 by immunohistochemistry. We observed many tumor-infiltrating CD45+ immune cells, F4/80+ macrophages, and CD3+ T cells in E0771 cell-derived tumors (data not shown). We further analyzed the tumor immune infiltrate in untreated E0771 by 17-color flow cytometry analysis. E0771 cell grafts were infiltrated mainly by TAMs (55.3% of all CD45+ cells), particularly M2-polarized cells (40.7% of all CD45+ cells), followed by NK cells (7.4% of all CD45+ cells), and CD4+ and CD8+ T cells (2.8% and 2.8% of all CD45+ cells, respectively) (data not shown), as observed in human TNBC (22). Direct comparison of the percentage of immune cell subpopulations among all living cells in the E0771 reinforced the findings that the E0771 cell- based model displayed strong immune cell infiltration, mainly by the myeloid and lymphoid compartments. These results suggested that the E0771 cell-based TNBC model could be defined as highly immunogenic with a potential anti-tumor effector response. Anti-Cath-D antibody-based therapy is effective in the immune-prone E0771 TNBC model First, we studied the antitumor effects of F1 and F1M1 in the immune-prone TNBC model (E0771 cells). We treated tumor-bearing mice with similar luciferase bioluminescence levels (data not shown), with F1, F1M1, or C1.18.4 isotype control (CTRL) (15 mg/kg, 3 times per week for 31 days; n=9 mice/group). F1 and F1M1 significantly delayed tumor growth compared with control (Fig.2A; P =0.005 for F1, P=0.002 for F1M1). Only 1/9 mice in the F1 group and 2/9 mice in the F1M1 group did not respond to treatment (Fig. 2B). At day 33 (sacrifice day), tumor volume was significantly reduced by 51.9% (P=0.0136) in the F1 group and by 61.9% (P=0.0194) in the F1M1 group compared with control (Fig.2C). Moreover, mice treated with F1 and F1M1 gained weight like the isotype control (Fig.2D) and displayed normal activities, suggesting no apparent toxicity. Drug-induced neutropenia is a potentially serious and life-threatening adverse event that may occur after therapy with various agents, including antibodies. Analysis by 17-color flow cytometry of blood from mice grafted with E0771 cells (data not shown) showed that neutrophil count at day 33 was not affected, indicating the absence of neutropenia (data not shown). Anti-Cath-D antibody-based therapy rebalances TAM recruitment and activation in tumors To study F1 and F1M1 immunomodulatory properties, we first analyzed by 17-color flow cytometry the immune infiltrates of E0771 cell grafts at treatment end. We focused on TAMs because they have been associated with TNBC progression and relapse (23). TAM (F4/80+ CD11b+ cells) percentage within the immune CD45+ cell population significantly decreased by 26.9% and 28.3% in F1- and in F1M1-treated animals (P=0.039 and P=0.046) compared with the C1.18.1 control (Fig.3A). Moreover, the proportion of F4/80+ CD206+ M2- polarized TAMs (with pro-tumorigenic functions) (6) within the immune F4/80+ CD11b+ TAM population significantly decreased by 33.4% and 43.5% in F1- and F1M1-treated animals (P=0.031 and P=0.011) compared with control (Fig.3B, left panel). Conversely, the proportion of F4/80+ CD206- CD11c+ M1-polarized TAM was comparable in all groups (Fig. 3C, left panel). However, the proportion of activated (CD86+) M1-polarized TAMs (capable of antigen presentation) increased by 163.3% and 153.9% in F1- and F1M1-treated animals (P=0.14 and P=0.049) compared with control (Fig. 3C, middle panel). Linear regression analysis showed that the percentages of M2-polarized TAMs and of CD86+ M1-polarized TAMs were correlated with tumor volume in all animals (three treatment groups together) (R2=0.1988, P=0.0290, and R2=0.2087, P=0.0248, respectively) (Fig. 3B and 3C, right panels). This suggested that the decreased tumor infiltration by M2-TAMs and activation of M1-TAMs may contribute to F1 and F1M1 anti-tumor effect. RT-qPCR analysis of tumor samples showed that the expression of the genes encoding chemokine ligand 2 (Ccl2) and its C-C motif chemokine receptor type 2 (Ccr2), which are involved in TAM recruitment, was significantly downregulated by 30.1% (P=0.0575) and 74.1% (P=0.0016) in the F1 group, and by 50.2% (P=0.0190) and 71.9% (P=0.0185) in the F1M1 group compared with control (Fig.3D, upper panels). Moreover, the expression level of Il10 (immunoregulatory cytokine, as a read-out of M2-polarized TAM immunosuppressive activity (23)), was significantly decreased by 68.2% (P=0.0031) and by 91.5% (P=0.0003) in the F1 and F1M1 groups, respectively. Similarly, expression of Mmp9 (matricellular protease associated with extracellular matrix remodeling and secreted by M2-polarized TAMs in BC was significantly downregulated by 66.6% (P=0.0031) and by 93.2% (P=0.0003) in the F1 and F1M1 groups, respectively (Fig.3D, bottom panels). Therefore, anti-Cath-D antibody-based therapy inhibited the recruitment of pro-tumor M2-polarized TAMs and increased the pool of activated anti-tumor M1-polarized TAMs in the E0771 TNBC preclinical model. Our data strongly suggest that in preclinical immunocompetent models of TNBC, anti-Cath-D antibody therapy modifies the myeloid immune population composition in the tumor microenvironment, leading to a less immunosuppressive microenvironment and the induction of the anti-tumor response. Anti-Cath-D antibody-based therapy induces NK cell activation and cytotoxic activity in tumors NK cells are frequently involved in the efficacy of antibody-based immunotherapies by triggering antibody-dependent cell-mediated cytotoxicity upon binding of the antibody fragment crystallizable (Fc) to Fcγ receptors (FcγR) on target immune cells. Therefore, we identified and quantified NK cells (i.e. CD45+ F4/80- CD3- Ly6C- Ly6G- CD11blo/+ NKp46+ cells) in E0771 cell grafts by 17-color flow cytometry at treatment end (day 33). The percentage of NK cells within the immune CD45+ cell population was comparable in the F1, F1M1, and control groups (Fig.4A, left panel). However, the percentage of tumor NKp46+ NK cells that expressed the activation marker CD16 (FcγRIII) was increased by 203.2% (P=0.0002) and by 172% (P=0.0142) in F1- and F1M1-treated animals (Fig.4A, right panel). RT-qPCR analysis showed that the gene encoding interleukin-15 (Il15), a cytokine associated with NK cell activation, was upregulated up to 652% (P=0.0109) and 325% (P=0.0939) in the F1 and F1M1 groups, respectively, compared with control (Fig. 4B, left panel). Similarly, Fcgr1, Fcgr2b, Fcgr3, Fcgr4 and Fcgrt (genes encoding FcγR involved in cell-mediated immunity) were highly upregulated by 4430% (P=0.0016), 5430% (P=0.0016), 2046% (P=0.0016), 2438% (P=0.0016), and 489.4% (P=0.0016) in the F1 group, and by 4255% (P=0.0003), 3030% (P=0.0003), 1580% (P=0.0003), 2276% (P=0.0003), and 908.6% (P=0.0003) in the F1M1 group compared with control (data not shown). Tnf (encoding an antitumor cytokine) was significantly upregulated by 416% (P=0.0295) and by 1580% (P=0.0003) in the F1 and F1M1 groups, respectively (Fig.4B, right panel). Conversely, in PBLs from E0771 cell-grafted mice, the percentages of NK cells and CD16+ NK cells were comparable among groups, suggesting that NK cells are specifically activated within tumors upon anti-Cath-D antibody therapy (data not shown). Altogether, our data demonstrated that anti-Cath-D antibody-based therapy induced NK cell activation and cytotoxic activity in tumors in preclinical immunocompetent models of BC. Anti-Cath-D antibody-based therapy triggers cDC1 cell recruitment and maturation in tumors To assess F1 and F1M1 effect on the recruitment and maturation status of cDCs, we quantified by 17-color flow cytometry cDCs (i.e. CD45+ CD3- Ly6G- B220+ CD11c+/hi cells), cDC2 (i.e. CD11c+/hi CD8- CD11b+ cells) and cDC1 cells (i.e. CD11c+/hi CD8+ CD11b- cells) in E0771 cell grafts at day 33. The percentages of tumor-recruited cDCs and cDC2 cells within the immune CD45+ cell population were comparable in the three groups. Conversely, the proportion of cDC1 cells was increased by 244% (P=0.0164) and by 311% (P=0.0055) in the F1 and F1M1 groups, respectively, compared with control (Fig.5A). Moreover, the proportion of tumor-infiltrating cDC1 cells that expressed the maturation surface marker CD86 was significantly increased by 750% (P=0.036) and by 500% (P=0.0053) in the F1 and F1M1 groups (Fig.5B), suggesting the setting/establishment of an adaptive antitumor response. cDC1 cells are implicated in antigen presentation to major histocompatibility (MHC) class I (MHC class I) molecules, leading to activation of cytotoxic responses by CD8+ T cells, and in the anti- tumor immunity in response to immunotherapie. The expression of H2d1, H2k1, and beta-2- microglobulin (B2m) (genes encoding proteins involved in antigen presentation via MHC class I molecules) was significantly upregulated by 8219% (P=0.0016), 2015% (P=0.0016) and 4561% (P=0.0016) in the F1 group, and by 13599% (P=0.0003), 3320% (P=0.0003) and 6693% (P=0.0003) in the F1M1 group compared with control (Fig. 5C). Similarly, the expression of IL12p40, which encodes interleukin-12 that is secreted by mature cDC1 cells and boosts the anti-tumor activity of T cells and NK cells within the tumor, was upregulated by 1358% (P=0.087) and by 11198% (P=0.0022) in the F1 and F1M1 groups, respectively (Fig. 5C). Moreover, the percentage of tumor-infiltrating CD86+ B cells, but not of all B cells recruited to the tumor, was significantly increased by 223.4% (P=0.0418) and by 232.8% (P=0.0152) in the F1 and F1M1 groups, respectively (data not shown). In conclusion, anti-Cath- D antibody therapy triggered cDC1 recruitment and maturation in tumors to potentially promote antigen presentation and the antitumor T-cell co-stimulation in preclinical immunocompetent models of BC. Anti-Cath-D antibody-based therapy reduces T-cell exhaustion in tumors and draining lymph nodes To analyze the recruitment and phenotype of tumor-infiltrating T cells in E0771 cell- grafted C57BL/6 mice in response to anti-Cath-D antibodies, we quantified by 17-color flow cytometry CD3+/CD4+ and CD3+/CD8+ T cells within the CD45+ immune cell population. The proportions of CD8+ and CD4+ T cells were comparable in the three groups (Fig. 6A-B, left panels). Immunotherapy targeting the PD-L1/PD-1 axis checkpoint blockade to rescue T cells from exhaustion has become an essential therapeutic strategy in cancers, including TNBC (4). Therefore, we studied PD-1 and PD-L1 expression on tumor-infiltrating CD4+ and CD8+ T cells. PD-L1 has recently been shown to be expressed by T cells, promoting immunosuppression in the tumor environment and is related to tumor immune tolerance and the adaptive immune response. The percentage of tumor-infiltrating CD8+ and CD4+ T cells that expressed the exhaustion marker PD-1 was comparable in the three groups (data not shown). Conversely, the percentage of PD-L1-expressing tumor-infiltrating CD4+ and CD8+ T cells was significantly decreased by 38.7% (P=0.0164) and 29.3% (P=0.0311) in the F1 group and by 38.5% (P=0.0206) and 35.1% (P=0.036) in the F1M1 group, compared with control (Fig. 6A-B, right panels). Similarly, in dLNs, the percentage of CD4+ T cells that expressed PD-L1 and lymphocyte-activation gene 3 (LAG3, another exhaustion marker) was decreased by 43.9% (P=0.0418) and 78.1% (P=0.0873) and by 29.2% (P=0.1996) and 64% (P=0.1919) in the F1 and F1M1 groups (data not shown). PD-L1+ and LAG3+ CD8+ T cells also were decreased by 49% (P=0.0115) and 53.5% (P=0.0079) (F1) and by 33.3% (P=0.0274) and 47.1% (P=0.0219) (F1M1) (data not shown), without any change in the overall T cell population (data not shown). Conversely, in PBLs, only the proportion of LAG3+ CD4+ T cells was decreased in the F1M1 group (data not shown). These results indicate T-cell exhaustion decreases in tumors and dLNs in E0771 cell-grafted immunocompetent mice upon anti-Cath-D antibody therapy. In agreement, the tumor expression levels of Lag3 and Tigit (T-cell immunoreceptor with Ig and ITIM domains), two genes involved in T-cell exhaustion (35), were reduced significantly by 67.3% (P=0.0109) and 74.5% (P=0.0016) in the F1 group, and by 89.3% (P=0.0006) and 79.5% (P=0.0012) in the F1M1 group, compared with control (Fig. 6C). Moreover, forkhead box P3 (Foxp3 encoding a Treg marker) and Il4 (supporting Treg-mediated immune suppression) were significantly downregulated in tumors by 65.9% (P=0.0016) and 66.4% (P=0.0016) in the F1 group, and by 74.2% (P=0.0129) and 49.8% (P=0.018) in the F1M1 group (Fig.6D). Collectively, our data highlight that anti-Cath-D antibodies restored the anti-tumor immunity by reducing T-cell exhaustion in tumors and in dLNs in E0771 cell grafts (preclinical model of TNBC). Our results also suggest that anti-Cath-D antibodies could reduce the tumor Treg pool in this model. EXAMPLE 2: F1M1-Fc+, a novel human Fc-engineered anti-cathepsin D antibody with enhanced ADCC and in vivo efficacy, triggers the recruitment and activation of natural killer cells and improves paclitaxel and enzalutamide therapy in triple-negative breast cancer Materials & Methods Binding of human Fc-engineered anti-cathepsin D antibodies to human CD16a (hCD16a) To analyze the anti-cath-D antibody binding to human FcγRIIIA (hCD16a), hCD16a 158V or 158F-transduced NK92 cells (2 × 104) were incubated with different concentrations of the F1M1-Fc-, F1M1, or F1M1-Fc+ antibodies. Then, cells were incubated with APC- conjugated 3G8 (anti-hCD16a antibody; 1:100) (4°C, 30min), washed twice in PBS at 4°C, and analyzed by flow cytometry. Fluorescence was acquired with a MACSQuant cytometer (Miltenyi) and analyzed with the Kaluza software version 2.1 (Beckman Coulter). Results were expressed as the percentage of the fluorescent 3G8 binding inhibition: (MFI without anti-cath- D antibody - MFI with anti-cath-D antibody) × 100/ (MFI without anti-cath-D antibody). This allowed the standardization and comparison between hCD16a 158V- and 158F-transduced NK92 cells. In vitro stimulation and functional responses of hCD16a-transduced NK92 cells For in vitro stimulation and functional responses of hCD16a-transduced NK92 cells, NUNC Maxisorp 96-well plates were directly sensitized with F1M1-Fc-, F1M1, F1M1-Fc+, or 3G8 (5 µg/ml) at 4°C for 12h, or pre-coated with the M2E8 anti-cath-D antibody in PBS (2500 ng/well), then incubated or not with increasing concentrations of cath-D (3-50 nM) and finally incubated with F1M1-Fc+ (5 µg/ml). After three washes with phosphate-buffered saline (PBS)/Tween-20 solution (PBS-T; 45 µl Tween-20 in 100 ml PBS), plates were saturated with 1% bovine serum albumin for 1 hour, then washed three times with PBS-T. NK92 hCD16a 158V or 158F cells (1 x 105) in 100 µl RPMI were then incubated on non-sensitized or sensitized plates in presence of the anti-CD107a-PC5 antibody (1:20) and 0.1 µg/ml BD GolgiPlug containing brefeldin A (BD Biosciences) at 37°C in 5% CO2 humidified air for 4h. Cells were then fixed and permeabilized using the BD Cytofix/Cytoperm Plus Kit (BD Biosciences) and stained with an anti-IFNγ-PE antibody (1:20) at 4°C for 30min to detect intracellular IFNγ. The proportions of responding NK cells (degranulating (CD107a+), IFNγ producing (IFNγ+) and cells exhibiting both responses (CD107a+ IFNγ+) were analyzed by flow cytometry with a Cytoflex S cytometer and the Kaluza 2.1 software (Beckman Coulter). NK cell-mediated ADCC towards TNBC cells and CAFs MDA-MB-231 cells (1 x 104 target; T) or hCAF1 cells (2 x 104 target; T) were plated in 96-well plates. After 24h, they were first incubated with F1M1-Fc+, F1M1, F1M1-Fc- or cetuximab (positive control) at 37 °C for 30min. Then, cells were incubated with NK92 hCD16a 158V, NK92 hCD16a 158F at an E:T ratio of 10:1, or human primary expanded NK cells (effector; E) at an E:T ratio of 3:1 at 37 °C for 24 h. MDA-MB-231 cell lysis (ADCC) or hCAF1 cell lysis was evaluated by measuring LDH release by bioluminescence with the Cytotox 96 Non-radioactive Cytotoxicity Assay (Promega). The percentage of specific lysis in each sample was determined using the following formula: percentage of specific lysis = (sample LDH release – [E cell + T cell] spontaneous LDH release)/(T cell maximum LDH release -T cell spontaneous LDH release) x 100. For ADCC in cell spheroids, target MDA-MB-231 cells (5 x 103) were seeded on Ultra Low Attachment 96-well plates (#7007, Corning). After 72h, MDA- MB-231 cell spheroids were first incubated with F1M1-Fc+, F1M1-Fc-, or cetuximab at 37 °C for 30min, and then with effector NK92 hCD16a+ 158V cells (E:T ratio of 20:1) for 24h. ADCC was assessed using the Cytotox 96 Non-radioactive Cytotoxicity Assay (Promega). The percentage of specific lysis in each sample was determined as described above. In vivo isolation and immunophenotyping of tumor-infiltrating NK cells Tumors were shredded and digested with a mixture of collagenase IV (1mg/mL) and DNase I (200 U/mL) in RPMI at 37°C. The mixture was transferred into a GentleMACS C tube for enzymatic and mechanical digestion using a gentleMACS™ Octo Dissociator (Miltenyi Biotec) at 37°C. After digestion, tumor suspensions were passed through a 70µm nylon cell strainer (#22-363-548, Thermo Fisher Scientific, Waltham, MA), centrifuged and resuspended in FACS Buffer (PBS pH 7.2, 1% decomplemented FCS, 2mM EDTA, and 0.02% sodium azide). Cells were blocked with FACS Buffer containing 1% (v/v) of mouse Fc block for 30min and stained with fluorescent-conjugated antibodies against the cell surface markers Viakrom, CD45, CD3, CD19, CD11c, F4/80, NKp46, CD27, CD11b, and CD107a for 45min. Then, cells were fixed, permeabilized and stained with fluorescent-conjugated antibodies against the intracellular marker granzyme B overnight. After fixation with 1% paraformaldehyde in PBS, cell samples were analyzed by flow cytometry using a Beckman and Coulter Cytoflex flow cytometer and FlowJo 10.8.1. Living immune cells were defined as Viakrome IR808- CD45+. NK cells were defined as NKp46+ cells within the gate excluding CD3+ T and NK T cells, CD19+ B cells, F4/80+ macrophages, and CD11c+ dendritic cells. NK cell maturation stage was analyzed with the CD27 and CD11b cell surface markers. Functional markers (CD107a, granzyme B) were studied in the NK cell subpopulations. Statistical analysis. A linear mixed regression model was used to determine the relationship between tumor growth and number of days post-xenograft. The variables included in the fixed part of the model were: number of days post-xenograft and treatment groups. Their interaction was also evaluated. Random intercepts and random slopes were included to take into account the time effect. The model coefficients were estimated by maximum likelihood. Survival rates were estimated using the Kaplan-Meier method and compared with the Log-rank test. Comparisons of more than two groups were done using the non-parametric Kruskal-Wallis test; the Mann- Whitney test was used for pairwise comparisons. Statistical significance was set at the 0.05 level. All statistical analyses were done with STATA 13.0. Results In TNBC, cath-D is a tumor microenvironment antigen eligible for Fc-engineered antibody targeted therapy to trigger ADCC ADCC initiation requires the presence of antibody-antigen complexes at the cell surface. Cath-D localizes at the cell surface binding to the M6P/IGF2 receptor via its mannose-6- phosphate (M6P) motif (BC cells and stromal fibroblasts) and to LRP1 (fibroblasts). We first examined the expression of cath-D, M6P/IGF2 receptor and LRP1 in a panel of TNBC cell lines (MDA-MB-231, SUM-159, MDA-MB-436, MDA-MB-453, BT-549) and human breast cancer-associated fibroblasts (hCAF1) cells (data not shown). We detected the fully mature cellular lysosomal protease cath-D (34-kDa heavy chain) and the IGF2/M6P receptor in all TNBC cell lines and hCAF1s. Conversely, LRP1 was weakly expressed by TNBC cells, except by SUM159 and BT549 cells, and well expressed by hCAF1 cells. The 52-kDa cath-D precursor was secreted in the culture medium of all TNBC cell lines and of hCAFs and was detectable in cell lysates, in agreement with its cell surface association after secretion (data not shown). Moreover, we detected the 52-kDa cath-D precursor and IGF2/M6P receptor in the cytosol (i.e. whole tumor lysate) of eight primary TNBC samples and in MDA-MB-231 and SUM-159 cell xenografts (data not shown). Immunofluorescence analysis of cath-D in MDA-MB-231 cell xenografts showed intense, punctate staining at the tumor periphery and in cell membrane protrusions, potentially in areas of exocytosis (data not shown), reflecting cath-D expression profile in TNBC biopsy samples (data not shown). This is in agreement with recent IHC results showing membrane-associated cath-D expression in 85.7% of TNBC samples in a tissue microarray. These observations strongly suggest that cath-D is a tumor microenvironment antigen eligible for Fc-engineered antibody targeted therapy to trigger ADCC. Generation of human IgG1 anti-cath-D F1M1 Fc-variant antibodies with different CD16a binding properties We next evaluated whether manipulation of the Fc portion of human IgG1 F1, our lead anti-cath-D antibody, by protein-engineering to increase ADCC could potentiate its antitumor effectiveness in TNBC. We first improved F1 format by introducing two-point mutations (one in VL CDR3 and one in VH FR3) to abrogate N-glycosylation at these two sites (F1M1 antibody) that may lead to the production of glycoforms associated with the risk of immunogenic responses in patients, as described for cetuximab. The migration profiles of F1 and F1M1 in the IgG1 format showed that the light and heavy chains molecular weights were lower in F1M1 than F1, as expected due to reduced glycosylation (data not shown). Sandwich ELISA using cath-D secreted from MDA-MB-231 cells showed that F1M1 (data not shown) had good binding capacities (0.04 nM), as previously shown for F1 (0.2 nM). Sandwich ELISA using cath-D secreted from mouse E0771 TNBC cells showed that F1M1 and F1 (Figure7A) retained good binding capacities also towards mouse cath-D (EC50=0.9 nM and 26.3 nM, respectively). The lower EC50 of F1M1 indicated that the two-point mutations leading to Fab aglycosylation enhanced F1M1 capacity to recognize human and mouse cath-D. To compare the anti-tumor efficacy of F1M1 and F1, we xenografted athymic Foxn1nu nude mice with MDA-MB-231 cells. When tumor volume reached 50 mm3, we treated mice with F1M1, F1, or negative isotype control (rituximab) (15mg/kg) by ip three times per week for 35 days (day 21- 56 post-graft), and then sacrificed all mice at treatment end. Both F1M1 and F1 significantly delayed tumor growth compared with negative control (data not shown; P=0.003 for F1M1, P<0.001 for F1). Thus, the VH and VL aglycosylated F1M1 antibody appears to be a suitable candidate for future clinical development. We then designed a human Fc-optimized F1M1 (F1M1-Fc+), in which the S239D, H268F, S324T, I332E mutations should increase the binding affinity to CD16a (FcγRIIIA) to enhance NK cell activation, and a F1M1 Fc-silent (F1M1-Fc-) in which the L234A, L235A and P329G mutations prevented binding to FcγRs and ADCC induction. F1M1-Fc- activities should rely exclusively on Fab-mediated effector functions. Antibody preparations were highly pure, and after SDS-PAGE in reducing conditions, the light chains were of the expected molecular mass (25-kDa) (data not shown). The three F1M1 antibody variants showed almost identical, concentration-dependent binding to secreted cath-D with EC50 values in the low nanomolar range (EC50=0.07nM, 0.051nM, and 0.048nM for F1M1, F1M1-Fc- and F1M1-Fc+, respectively) (figure 7B). Moreover, F1M1-Fc+ binding to pro-cath-D was comparable at pH values from 7.2 to 5.6 (data not shown). All F1M1 variant antibodies immunoprecipitated secreted cath-D at pH values from 7.5 to 5.5 (data not shown), suggesting that they were all active in the highly acidic tumor microenvironment. Then, we investigated by surface plasmon resonance F1M1-Fc+, F1M1 and F1M1-Fc- binding to hCD16a (158V or 158F allotype). F1M1 binding to hCD16a 158V was 3.9-fold higher than to hCD16a 158F (KD= 42 nM and KD 164 nM, respectively) (Figure 7C). Compared with F1M1, F1M1-Fc+ binding to hCD16a 158V was increased by 21.2-fold (KD=2 nM for F1M1-Fc+ and 42.4 nM for F1M1), and even more to hCD16a 158F (by 54.6-fold) (KD=3 nM for F1M1-Fc+, and 164 nM F1M1). As F1M1-Fc- showed no detectable binding to both hCD16a allotypes, we used it as control devoid of effector function. Finally, we investigated by flow cytometry F1M1-Fc+, F1M1 and F1M1-Fc- binding to hCD16a (158V or 158F allotype) expressed on the surface of human NK92 cells. It has been shown that the hCD16a 158V/F polymorphism in NK cells influences its binding to human IgG131. Specifically, hCD16a V158 displays increased affinity for human IgG1, resulting in increased NK cell activation. F1M1 binding to hCD16a 158V was approximately 5-fold higher than to hCD16a 158F at all tested concentrations (Figure 7G). Compared with F1M1, F1M1-Fc+ binding to hCD16a 158V was increased (2- to 3-fold), and even more to hCD16a 158F (7- to 20-fold). Engineered human F1M1-Fc+ increases NK cell activation in vitro, and ADCC of TNBC cells and CAFs We next compared F1M1-Fc+, F1M1 and F1M1-Fc- capacity to activate NK cells by analyzing cell surface CD107a expression, as a functional marker of NK cell degranulation, and intracellular IFNγ, a cytokine secreted following NK cell activation, in hCD16a-transduced human NK92 cells. The percentages of CD107a+ NK, IFNγ+ NK and CD107a+ IFNγ+ NK subsets were increased similarly by 2- to 3-fold following incubation with F1M1-Fc+, compared with F1M1, in hCD16a 158V- or 158F-expressing NK cells (figure 8A). Conversely, F1M1- Fc- did not activate NK effector functions. Moreover, the percentages of CD107a+ NK, IFNγ+ NK and CD107a+ IFNγ+ NK cells were increased in a dose-dependent manner in the presence of increasing concentrations of cath-D bound to F1M1-Fc+ (figure 8B). Overall, F1M1-Fc+ was the most potent antibody to activate NK cell response. Next, we determined whether the F1M1- Fc+-enhanced binding to hCD16a-expressing NK cells translated into increased cytotoxic activity. First, incubation of MDA-MB-231 cells with F1M1-Fc+ significantly increased ADCC by 5-fold (P<0.0001) and 18-fold (P=0.0002), compared with F1M1, in the presence of NK92- hCD16a 158V cells and NK92-hCD16a 158F cells, respectively (figure 8C). F1M1-Fc- did not induce ADCC. Moreover, F1M1-Fc+ triggered ADCC in a dose-dependent manner in the presence of NK92-hCD16a 158V cells (figure 8D). F1M1-Fc+-induced ADCC was exclusively dependent on Fc binding to the hCD16 receptor on NK92-hCD16a 158V cells, because addition of an Fc block completely inhibited ADCC, to a level similar to that observed with F1M1-Fc- (figure 8E). We then evaluated ADCC against MDA-MB-231 cells using primary human NK cells (158V/F allotype) (data not shown). ADCC was significantly increased by 3.5-fold upon incubation with F1M1-Fc+, compared with F1M1 (P<0.0001) (data not shown). As cath-D localizes at the surface of BC cells and stromal fibroblasts by binding to the M6P/IGF2 receptor, we next asked whether M6P/IGF2 receptor-bound cath-D was involved in ADCC induction by F1M1-Fc+. Thus, we evaluated ADCC against MDA-MB-231 cells in the presence of NK92-hCD16a 158V cells and of excess M6P to compete with cath-D binding to the M6P/IGF2 receptor (figure 8F). Addition of M6P, but not glucose-6-phosphate (negative control for M6P), significantly inhibited ADCC induction by F1M1-Fc+ by 51% (P<0.0001), indicating that cath-D bound to its M6P/IGF2 receptor at the cell surface was involved in ADCC. Similarly, using primary human NK cells (158 V/V allotype) (data not shown), ADCC of MDA-MB-231 cells was inhibited by 70% (P<0.0001) in the presence of M6P (data not shown). Moreover, F1M1-Fc+ efficiently triggered ADCC against hCAF1 cells in the presence of NK92-hCD16a 158V cells, and excess M6P significantly inhibited ADCC by 62% (P<0.0001) (figure 8G). Lastly, to better recapitulate the in vivo cancer cell environment, we prepared spheroids of MDA-MB-231 cells co-cultured with NK92 hCD16a 158V cells. F1M1- Fc+ also induced ADCC of MDA-MB-231 cell spheroids (figure 8H). Live imaging of ADCC in calcein-labeled MDA-MB-231 cell spheroids showed their time-dependent lysis in response to F1M1-Fc+ exposure (data not shown). At 24h, the fluorescence signal of calcein-labeled MDA-MB-231 spheroids was significantly decreased by 29.5% in response to F1M1-Fc+ compared with F1M1-Fc- (P=0.005) (data not shown). Overall, our results demonstrated that F1M1-Fc+ is the most potent anti-cath-D antibody to trigger ADCC in TNBC cells and CAFs. F1M1-Fc+ is the best candidate to reduce MDA-MB-231 cell xenograft growth and improve nude mouse survival. To compare the in vivo anti-tumor efficacy of F1M1-Fc+, F1M1, and F1M1-Fc-, we used athymic Foxn1nu nude mice subcutaneously xenografted with MDA-MB-231 cells. When MDA-MB-231 tumors reached 50 mm3, we treated mice with F1M1-Fc+, F1M1, F1M1-Fc-, or the anti-human CD20 IgG1 rituximab, as negative isotype control (Ctrl) (15mg/kg) by ip three times per week for 35 days (day 13-48 post-graft), and sacrificed mice when tumor volume reached 2000 mm3. Both F1M1-Fc+ and F1M1 significantly delayed tumor growth compared with negative control (P=0.001 for F1M1-Fc+, P=0.011 for F1M1). F1M1-Fc- slightly delayed tumor growth (P=0.057) (figure 9A). F1M1-Fc+ tended to be more effective than F1M1-Fc- (P=0.055). At day 48 (treatment end), tumor volume was significantly reduced by 77% (P=0.0002) in the F1M1-Fc+ group, by 60% (P=0.0007) in the F1M1 group, and by 40.5% (P=0.0289) in the F1M1-Fc- group compared with control (figure 9B). Tumor volume was significantly smaller in the F1M1-Fc+ group than in the F1M1-Fc- group (P=0.0041). The overall survival rate, reflected by a tumor volume <2000 mm3, was significantly longer in the F1M1-Fc+, F1M1, and F1M1-Fc- groups than in controls. The median survival was 59, 57, 54.5, and 50 days in the F1M1-Fc+, F1M1, F1M1-Fc-, and control groups (figure 9C; Kaplan-Meier survival analysis, P=0.0003 for F1M1-Fc+, P=0.0011 for F1M1, P=0.0275 for F1M1-Fc- compared with control). The overall survival rate was significantly longer in mice treated with F1M1-Fc+ than with F1M1-Fc- (P=0.0253). Moreover, all mice treated gained weight (figure 9D) and displayed normal activities, suggesting no apparent toxicity. F1M1-Fc+ triggers in vivo recruitment and activation of NK cells to induce ADCC in MDA-MB-231 TNBC cell xenografts. To investigate the in vivo mechanisms underlying the antitumor effect of F1M1-Fc+, F1M1, and F1M1-Fc-, we treated nude mice xenografted with MDA-MB-231 cells with F1M1- Fc+, F1M1, F1M1-Fc-, or rituximab as negative isotype control (same schedule as before), and then sacrificed all mice at treatment end (day 48). F1M1-Fc+, F1M1, and F1M1-Fc- inhibited tumor growth compared with rituximab, but only F1M1-Fc+ resulted in significant tumor growth inhibition compared with rituximab (P=0.003) (figure 10A). At day 48, tumor volume was significantly reduced by 60.3% (P=0.0005) in the F1M1-Fc+ group, by 42.9% (P=0.020) in the F1M1 group, and by 36.6% (P=0.036) in the F1M1-Fc- group compared with control group (figure 10B). Tumor volume was significantly more reduced in the F1M1-Fc+ group than in the F1M1-Fc- group (P=0.0368). Human cath-D expression level in tumors was not affected by F1M1-Fc+, F1M1, or F1M1-Fc- treatment, as shown by IHC and western blot analyses (figure 11A-B). To study the impact of the Fc part of F1M1 on NK cell recruitment and activation, we identified and quantified by flow cytometry tumor-infiltrating NK cells in MDA-MB-231 xenografts after treatment end. The overall percentage of living cells was not different between untreated (control) and treated groups (data not shown). The percentage of NK (CD45+ F4/80- CD3- CD19- CD11c- NKp46+) cells within the living immune IR808-CD45+ cell population was significantly increased by 207.2% in the F1M1-Fc+ group compared with control (P=0.0053), but not in the F1M1- and F1M1-Fc- groups (figure 10C). Linear regression analysis showed that the NK cell percentage was inversely correlated with tumor volume in all animals (four treatment groups together) (R2=0.2139, P=0.0062) (data not shown). This suggested that in the F1M1-Fc+ group, NK cell recruitment in the tumor contributed to the anti-tumor response. We also determined the NK cell phenotype based on the expression of the NK cell surface maturation markers CD11b and CD27. Only the CD27CD11b NK cell population was significantly increased by 268% in F1M1-Fc+-treated animals (P=0.0016 compared with the control group), but not in the F1M1- and F1M1-Fc--treated groups (data not shown). This suggested that F1M1-Fc+ treatment resulted in the recruitment mainly of immature NK cells. The CD27+CD11b- NK cell population (mature stage 1) also tended to be increased in the F1M1-Fc+ group by 222.5% (P=0.19 compared with the control group) (data not shown). NK cell development and maturation is orchestrated by a network of transcription factors, including Eomes. Eomes is mainly expressed in immature NK cells, promotes survival of maturing NK cells, and plays a major role in the induction of genes associated with NK cell cytotoxicity, such as Prf1. RT-qPCR analysis of tumor samples showed that Eomes expression was significantly upregulated by 479.3% in the F1M1-Fc+ group (P=0.0006 compared with control), but not in the F1M1 and F1M1-Fc- groups (figure 10D). This strongly suggested that F1M1-Fc+-based therapy triggers the recruitment of immature NK cells that might become cytotoxic in the MDA- MB-231 TNBC model. Next, we analyzed cell surface expression of CD107a, a functional marker of NK cell degranulation, by flow cytometry of tumor-infiltrating NKp46+ cells in MDA-MB-231 cell xenografts at day 48. The percentage of CD107a+ NK cells within the NK cell population was significantly increased by 142% in F1M1-Fc+-treated animals (P=0.0464 compared with control), but not in the F1M1- and F1M1-Fc- groups (figure 10E). The percentage of CD107a+ NK cells was higher (by 142.6%) in the F1M1-Fc+ group compared with the F1M1-Fc- group (P=0.0079). Linear regression analysis showed that the percentage of CD107a+ NK cells was inversely correlated with tumor volume in all animals (four treatment groups together) (R2=0.1703, P=0.0211) (data not shown). This suggested that in the F1M1-Fc+ group, in vivo tumor infiltration by activated CD107a+ NK cells may contribute to the anti-tumor response. Lastly, we analyzed intracellular granzyme B (GZMB) expression as a marker of NK cell cytotoxic activity by flow cytometry in tumor-infiltrating NKp46+ cells in MDA-MB-231 cell xenografts at day 48. The percentage of GZMB+ NK cells was significantly increased by 173.8% (P=0.0079) and by 185.4% (P=0.0289) in the F1M1-Fc+ and F1M1 groups, respectively, compared with control, but not in the F1M1-Fc- group (figure 10F). It also was higher (by 193%) in the F1M1-Fc+ group than in the F1M1-Fc- group (P=0.0418). Linear regression analysis showed that the percentage of GZMB+ NK cells was inversely correlated with tumor volume in all animals (four treatment groups together) (R2=0.3085, P=0.0012) (data not shown). This suggested that in the F1M1-Fc+ and F1M1 groups, tumor infiltration by NK cells with cytotoxic activity contributed to the anti-tumor response. In agreement, granzyme B (Gzmb) and perforin (Prf1) mRNA levels, as a read-out of NK cell cytotoxic activity, were upregulated (up to 166.3% and 173.8%,, respectively) in the F1M1-Fc+ group compared with control group (P= 0.3969 and P=0.2319), and also compared with the F1M1-Fc- group (up to 256.3% and 243.4%; P=0.0207 and P=0.0379, respectively) (data not shown). The antitumor cytokine Tnf secreted by activated NK cells, also was upregulated by 171.5% (P=0.0059) in the F1M1-Fc+ group and by 160.3% (P=0.0541) in the F1M1 group compared with control (data not shown). Tnf was upregulated (by 266.8%) (P=0.003) in the F1M1-Fc+ group compared with the F1M1-Fc- group. Altogether, these results demonstrated that F1M1-Fc+ triggers NK cell recruitment, activation and cytotoxic activity in MDA-MB-231 cell xenografts, and strongly suggest the key role of the Fc part of this anti-cath- D antibody in its anti-tumor activity via NK cells. NK cell depletion impairs F1M1-Fc+ therapeutic efficacy in MDA-MB-231 cell xenografts. The significant antitumor effects of F1M1-Fc+ and the F1M1-Fc+-mediated induction of NK cell recruitment, activation and cytotoxic activity strongly suggested that NK cells are crucial for F1M1-Fc+-antitumor activity. To confirm this hypothesis, we depleted NK cells in MDA-MB-231-bearing nude mice by intraperitoneal injection of an anti-asialo-GM1 antibody (αGM1). First, to determine the efficiency of NK cell depletion, we treated non-grafted nude mice with control (saline solution) or αGM1 at day 0 and day 3 (data not shown). Analysis of blood samples at day 4 and day 7 confirmed the efficacy of NK cell depletion, even four days (i.e. day 7) after the last αGM1 injection (data not shown). Other immune cells (B cells, neutrophils, dendritic cells and macrophages) were not affected by αGM1 treatment, as indicated by their quantification in blood and spleen (data not shown). Then, to investigate NK cell role in the antitumor effect of F1M1-Fc+, we treated nude mice harboring MDA-MB-231 cell xenografts with F1M1-Fc+ or rituximab (negative isotype control) in the presence or absence of αGM1, and then sacrificed all mice at treatment end (day 48), according to the schedule described in figure 12A. As previously observed, F1M1-Fc+ significantly inhibited tumor growth compared with control (P<0.001) (figure 12B). In the presence of αGM1, F1M1-Fc+ antitumor activity (tumor growth reduction) was decreased (P=0.021 versus control, and P=0.015 versus F1M1-Fc+ alone), revealing the crucial role of NK cells (Figure 14B). At day 48, compared with the control group, tumor volume was significantly reduced by 63.3% (P=0.0003) in the F1M1-Fc+ group, but only by 30.7% (P=0.0881) in the F1M1-Fc+ + αGM1 group (figure 12C). Moreover, tumor volume was significantly smaller in the F1M1-Fc+ group than in the F1M1-Fc+ + αGM1 group (P=0.0281) (figure 12C). The number of tumor-infiltrating NK cells (CD45+ CD3- CD19- Ly6G- NKp46+) within the living immune CD45+ cell population in tumors was significantly increased by 248.7% in the F1M1- Fc+ group compared with control (P=0.0312). Conversely, very few NK cells were detected in the F1M1-Fc+ + αGM1 group (figure 12D). Analysis of blood samples at day 45 and of the spleens at day 48 validated NK cell depletion in the F1M1-Fc+ + αGM1 group (data not shown). This demonstrated the key role of NK cells in F1M1-Fc+ therapeutic efficiency. Drug-induced neutropenia is a potentially serious and life-threatening adverse event that may occur after therapy with various agents, including antibodies (e.g. immune checkpoint inhibitors). Analysis by flow cytometry of blood from mice from the control and F1M1-Fc+ treated groups (from Figure 12) showed that neutrophil count at day 45 was not affected, indicating the absence of neutropenia (Figure 13A and 13B). Similarly, no sign of leukopenia, thrombopenia and anemia was observed after F1M1-Fc+ treatment, suggesting the absence of toxicity (data not shown). F1M1-Fc+ inhibits tumor growth of patient-derived TNBC xenografts and improves mouse survival. Next, we tested F1M1-Fc+ therapeutic effect in mice harboring TNBC-PDXs (B1995 and B3977). First, we analyzed the expression of cath-D and the M6P/IGF2 receptor in PDX B1995 and B3977 xenografts (the two fastest growing PDX in nude mice among a PDX collection), and in MDA-MB-231 and SUM-159 cell xenografts by western blot analysis. Cath- D and the M6P/IGF2 receptor were similarly expressed in all samples (data not shown). Immunostaining of the PDX B1995 and B3977 confirmed that cath-D was expressed in tumor cells and microenvironment (data not shown), as previously observed in TNBC biopsy samples (data not shown). These results indicated these PDX are representative of the disease, at least concerning cath-D expression. We then xenografted Swiss nude mice with the PDX B1995 or B3977. Tumor growth was significantly reduced by F1M1-Fc+ (15mg/kg by ip three times per week) compared with control (rituximab) in both models (P<0.001) (figure 11A-B, left panel). At treatment end (day 48 for the PDX B1995 and day 42 for the PDX B3977), tumor volume was significantly reduced by 61.8% (P=0.0002) and by 44.6% (P=0.0018), respectively, in the F1M1-Fc+ group compared with the control group (figure 14A-B, middle panels). The overall survival rate, reflected by a tumor volume <1500 mm3, was significantly longer in mice treated with F1M1-Fc+ than in control group (median survival of 64 and 49 days for the F1M1-Fc+ group and for control animals in mice xenografted with PDX B1995, and of 57 and 46 days for the F1M1-Fc+ group and for control animals in mice xenografted with PDX B3977) (figure 14A-B, right panel; Kaplan-Meier survival analysis, P=0.0002 and P=0.0024, respectively). These results showed that F1M1-Fc+ monotherapy very efficiently delayed tumor growth in nude mice xenografted with TNBC PDXs. F1M1-Fc+ improves the therapeutic efficacy of the microtubule inhibitor paclitaxel Next, we investigated the therapeutic benefit of combining F1M1-Fc+ with paclitaxel. First, we confirmed paclitaxel cytotoxicity in MDA-MB-231 cells in vitro (IC50 = 53.4 nM) (data not shown). Then, we showed that incubation of MDA-MB-231 cells with 5 nM paclitaxel for 48h (dose equivalent to the IC20; data not shown) did not affect cath-D expression (cell lysates) and secretion (conditioned media) (data not shown). As paclitaxel is frequently administered to patients with a weekly schedule, we treated nude mice bearing MDA-MB-231 cell xenografts with 1 mg/kg, 4 mg/kg, or 7 mg/kg paclitaxel (PTX) weekly. Higher PTX doses resulted in excessive toxicity (data not shown). PTX induced tumor growth inhibition in a dose- dependent manner (data not shown) that was significant at 4 mg/kg (P=0.007) and 7 mg/kg (P<0.001) compared with control (NaCl). We then tested the combination therapy (paclitaxel and F1M1-Fc+) in nude mice bearing MDA-MB-231 cell xenografts using a low dose (PTX LD, 1 mg/kg) and a medium dose of paclitaxel (PTX MD, 4 mg/kg that inhibited tumor growth on its own; data not shown). When tumor volume reached 50 mm3 at day 15 post-graft, we treated mice with F1M1-Fc+ (15mg/kg; three times per week), PTX LD (1 mg/kg; once per week), F1M1-Fc+ (15mg/kg; three times per week) + PTX LD (1 mg/kg; once per week), or control (rituximab, three times per week + saline, once per week; all ip) for 37 days. Both F1M1-Fc+ and PTX LD+F1M1-Fc+ significantly delayed tumor growth compared with control (P=0.039 for F1M1-Fc+, P=0.032 for PTX LD+F1M1-Fc+) (figure 15A). At day 52 (treatment end), tumor volume was reduced by 37.4% (P=0.07) in the F1M1-Fc+ group, by 18.7% (P=0.0005) in the PTX LD group, and by 50.5% (P=0.002) in the PTX LD+F1M1-Fc+ group compared with control group (figure 12B). Tumor volume was significantly smaller in the PTX LD+F1M1-Fc+ group than in the PTX LD group (P=0.0293). The overall survival rate, reflected by a tumor volume <2000 mm3, was significantly longer in mice treated with F1M1-Fc+ and with PTX LD+F1M1-Fc+ than in controls (median survival of 59 and 66 days for the F1M1-Fc+ and PTX LD+F1M1-Fc+ groups, respectively, compared with 55 days for control animals) (figure 15C; Kaplan-Meier survival analysis, P=0.0313 for F1M1-Fc+, P=0.0009 for PTX LD+F1M1-Fc+). The overall survival rate was significantly longer in mice treated with PTX LD+F1M1-Fc+ (66 days) than in mice treated with PTX LD (57 days) (P=0.0219). Overall, these results demonstrated that in combination therapy, F1M1-Fc+ improved the therapeutic efficacy of a suboptimal dose of paclitaxel that was poorly effective in vivo (data not shown). Similarly, F1M1-Fc+, PTX MD, and PTX MD+F1M1-Fc+ significantly delayed tumor growth compared with negative control (P=0.039 for F1M1-Fc+, P=0.042 for PTX MD, P=0.013 for PTX MD+F1M1-Fc+) (figure 15D). At day 52 (treatment end), tumor volume was reduced by 37.4% (P=0.07) in the F1M1-Fc+ group, by 32.9% (P=0.0289) in the PTX MD group, and by 55.5% (P=0.0012) in the PTX MD+F1M1-Fc+ group compared with control (figure 12E). The overall survival rate was significantly longer in mice treated with F1M1-Fc+, PTX MD and PTX MD+F1M1-Fc+ than in control (median survival of 59, 59 and 69 days for the F1M1-Fc+, PTX MD, and PTX MD+F1M1-Fc+ groups, respectively, compared with 55 days for control animals) (figure 12F; Kaplan-Meier survival analysis, P=0.0313 for F1M1- Fc+, P=0.0312 for PTX MD, P=0.001 for PTX MD+F1M1-Fc+). The overall survival rate was significantly longer in mice treated with PTX MD + F1M1-Fc+ (69 days) than with PTX MD (59 days) (P=0.041). All mice treated with the combination therapy gained weight (data not shown) and displayed normal activities, suggesting no apparent off-target effects. Overall, these results demonstrated that F1M1-Fc+ improved PTX therapeutic efficacy. F1M1-Fc+ improves the therapeutic efficacy of the anti-androgen enzalutamide We previously found that co-expression of cath-D and androgen receptor (AR) in non- metastatic TNBC is an independent prognostic factor of worse overall survival, suggesting the possibility of adjuvant combination therapy with anti-androgen drugs and anti-cath-D antibodies. We first showed that in the AR-expressing SUM159 TNBC cell line (data not shown), incubation with enzalutamide, an antiandrogen drug, did not decrease cath-D expression and secretion (data not shown), and incubation with F1M1-Fc+ did not reduce AR expression (data not shown). Then, we treated mice harboring SUM159 cell xenografts with F1M1-Fc+ alone. Tumor volume increase was significantly slowed down in mice treated with F1M1-Fc+ (15mg/kg by ip three times per week) compared with control (rituximab) (P<0.001) (figure 16A). Tumor volume at day 48 (treatment end) was significantly reduced by 71.9% in the F1M1-Fc+ compared with control group (P <0.0001) (figure 16B). The overall survival rate, reflected by a tumor volume <1000 mm3, was significantly longer in mice treated with F1M1-Fc+ than in the control group (median survival of 62 days and 55 days, respectively) (figure 16C; Kaplan- Meier survival analysis, P=0.0024), without any apparent toxicity (figure 16D). We then treated nude mice bearing SUM159 cell xenografts with F1M1-Fc+ (15mg/kg, twice per week, ip) or/and enzalutamide (Enza, 30mg/kg, five times per week, per os). F1M1- Fc+ and Enza+F1M1-Fc+ significantly delayed tumor growth compared with control (P=0.001 for F1M1-Fc+, P=0.003 for Enza+ F1M1-Fc+) (figure 16E). Tumor growth was significantly more reduced in the Enza+F1M1-Fc+ group than in the Enza group (P=0.008). At day 50 (treatment end), tumor volume was reduced by 55.7% (P=0.0019) in the F1M1-Fc+ group, by 43.4% (P=0.02) in the Enza group, and by 71% (P=0.0002) in the Enza+F1M1-Fc+ group compared with control (figure 16F). Tumor volume was significantly smaller in the Enza+F1M1-Fc+ group than in the Enza group (P=0.0082). Overall survival was significantly longer in mice treated with F1M1-Fc+, Enza, and Enza+F1M1-Fc+ than in controls (median survival of 63, 61, 64, and 57 days, respectively) (figure 16G; Kaplan-Meier survival analysis, P=0.0126 for F1M1-Fc+, P=0.0196 for Enza, P<0.0001 for Enza + F1M1-Fc+), without apparent toxicity (figure 16H). Overall survival was significantly longer in the Enza+F1M1- Fc+ group than in the Enza group (64 vs 61 days, P=0.017) and in the F1M1-Fc+ group (64 vs 63 days) (P=0.0091) (figure 16G). Overall, these results demonstrated that F1M1-Fc+ improves enzalutamide therapeutic efficacy. EXAMPLE 3: F1M1-ADCs inhibit tumor growth of 4-hydroxytamoxifen-resistant LCC2-MCF7 breast cancer cells and improve mouse survivalF1M1-MMAF were generated by bioconjugating F1M1 anti-cath-D via some of the eight cysteines forming interchain disulfide bridges, to auristatin F (MMAF) through a non-cleavable linker. F1M1-MMAE were generated by bioconjugating F1M1 anti-cath-D via some of the eight cysteines forming interchain disulfide bridges, to auristatin E (MMAE) through a through a valine-citrulline cleavable linker. We show that both F1M1-MMAF and F1M1-MMAE is able to inhibit tumor growth of Tam-resistant LCC2 BC cells and improve mouse survival (Figure 17A-17G) REFERENCES: Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure. 1. Bianchini G, De Angelis C, Licata L, Gianni L. Treatment landscape of triple-negative breast cancer - expanded options, evolving needs. Nat Rev Clin Oncol 2022;19(2):91-113 doi 10.1038/s41571-021-00565-2. 2. Wojtukiewicz MZ, Pogorzelska M, Politynska B. Immunotherapy for triple negative breast cancer: the end of the beginning or the beginning of the end? Cancer Metastasis Rev 2022;41(3):465-9 doi 10.1007/s10555-022-10060-4. 3. Barroso-Sousa R, Jain E, Cohen O, Kim D, Buendia-Buendia J, Winer E, et al. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann Oncol 2020;31(3):387-94 doi 10.1016/j.annonc.2019.11.010. 4. Cortes J, Rugo HS, Cescon DW, Im SA, Yusof MM, Gallardo C, et al. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N Engl J Med 2022;387(3):217-26 doi 10.1056/NEJMoa2202809. 5. Kyriazoglou A, Kaparelou M, Goumas G, Liontos M, Zakopoulou R, Zografos E, et al. Immunotherapy in HER2-Positive Breast Cancer: A Systematic Review. Breast Care (Basel) 2022;17(1):63-70 doi 10.1159/000514860. 6. Tariq M, Zhang J, Liang G, Ding L, He Q, Yang B. Macrophage Polarization: Anti- Cancer Strategies to Target Tumor-Associated Macrophage in Breast Cancer. J Cell Biochem 2017;118(9):2484-501 doi 10.1002/jcb.25895. 7. Jiang K, Dong M, Li C, Sheng J. Unraveling Heterogeneity of Tumor Cells and Microenvironment and Its Clinical Implications for Triple Negative Breast Cancer. Front Oncol 2021;11:557477 doi 10.3389/fonc.2021.557477. 8. Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 2017 doi 10.1016/S1470-2045(17)30904-X. 9. Souza-Fonseca-Guimaraes F. NK cell-based immunotherapies: awakening the innate anti- cancer response. Discov Med.2016;21(115):197-203. 10. Lopez-Soto A, Gonzalez S, Smyth MJ, et al. Control of Metastasis by NK Cells. Cancer Cell.2017;32(2):135-154. doi: 10.1016/j.ccell.2017.06.009. 11. Rugo HS, Im SA, Cardoso F, et al. Efficacy of Margetuximab vs Trastuzumab in Patients With Pretreated ERBB2-Positive Advanced Breast Cancer: A Phase 3 Randomized Clinical Trial. JAMA Oncol.2021;7(4):573-584. doi: 10.1001/jamaoncol.2020.7932. 12. Correia AL, Guimaraes JC, Auf der Maur P, et al. Hepatic stellate cells suppress NK cell- sustained breast cancer dormancy. Nature.2021;594(7864):566-571. doi: 10.1038/s41586-021- 03614-z. 13. Liu R, Oldham RJ, Teal E, et al. Fc-Engineering for Modulated Effector Functions- Improving Antibodies for Cancer Treatment. Antibodies (Basel). 2020;9(4). doi: 10.3390/antib9040064. 14. Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer. 2012;12(4):278-87. doi: 10.1038/nrc3236. 15. Trinchieri G, Valiante N. Receptors for the Fc fragment of IgG on natural killer cells. Nat Immun.1993;12(4-5):218-34. 16. Voskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol.2015;15(6):388-400. doi: 10.1038/nri3839. 17. Prager I, Watzl C. Mechanisms of natural killer cell-mediated cellular cytotoxicity. J Leukoc Biol.2019;105(6):1319-1329. doi: 10.1002/JLB.MR0718-269R. 18. Warren HS, Kinnear BF. Quantitative analysis of the effect of CD16 ligation on human NK cell proliferation. J Immunol.1999;162(2):735-42. 19. Kang J, Yu Y, Jeong S, Lee H, Heo HJ, Park JJ, et al. Prognostic role of high cathepsin D expression in breast cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 2020;12:1758835920927838 doi 10.1177/1758835920927838. 20. Mansouri H, Alcaraz LB, Mollevi C, Mallavialle A, Jacot W, Boissiere-Michot F, et al. Co-Expression of Androgen Receptor and Cathepsin D Defines a Triple-Negative Breast Cancer Subgroup with Poorer Overall Survival. Cancers (Basel) 2020;12(5) doi 10.3390/cancers12051244. 21. Vignon F, Capony F, Chambon M, Freiss G, Garcia M, Rochefort H. Autocrine growth stimulation of the MCF 7 breast cancer cells by the estrogen-regulated 52 K protein. Endocrinology 1986;118(4):1537-45. 22. Ashraf Y, Mansouri H, Laurent-Matha V, Alcaraz LB, Roger P, Guiu S, et al. Immunotherapy of triple-negative breast cancer with cathepsin D-targeting antibodies. J Immunother Cancer 2019;7(1):29 doi 10.1186/s40425-019-0498-z. 23. Wang S, Yang Y, Ma P, Zha Y, Zhang J, Lei A, Li N. CAR-macrophage: An extensive immune enhancer to fight cancer. EBioMedicine.2022 Feb;76:103873.

Claims

CLAIMS: 1. An isolated anti-Cathepsin-D antibody comprising (F1M1): (c) a heavy chain wherein the variable domain comprises: a H-CDR1 having a sequence set forth as SEQ ID NO: 2; a H-CDR2 having a sequence set forth as SEQ ID NO: 3; a H-CDR3 having a sequence set forth as SEQ ID NO: 4; (d) a light chain wherein the variable domain comprises : a L-CDR1 having a sequence set forth as SEQ ID NO: 6; a L-CDR2 having a sequence set forth as YDS; a L- CDR3 having a sequence set forth as SEQ ID NO: 7.
2. The isolated anti-Cathepsin-D antibody of claim 1, comprising: (a) a heavy chain wherein the variable domain has at least 70% of identity with a sequence set forth as SEQ ID NO:1, and (b) a light chain wherein the variable domain has at least 70% of identity with a sequence set forth as SEQ ID NO:5.
3. The isolated anti-Cathepsin-D antibody of claim 1 or 2, comprising (a) a heavy chain wherein the variable domain has a sequence set forth as SEQ ID NO:1 and (b) a light chain has a heavy chain wherein the variable domain has a sequence set forth as SEQ ID NO:5.
4. The isolated anti-Cathepsin-D antibody of any one of claims 1 to 3, wherein said antibody is a monoclonal antibody, and more particularly a monoclonal IgG antibody.
5. The isolated anti-Cathepsin-D antibody of any one of claims 1 to 4, wherein said antibody is selected from the group consisting of Fab, F(ab')2, Fab', dsFv, diabodies and scFv.
6. The isolated anti-Cathepsin-D antibody of any one of claims 1 to 4, wherein said antibody comprises a Fc region comprising at least 1, 2, 3 or 4 mutations selected from the group consisting of S239D, H268F, S324T and I332E (“F1M1-Fc+”).
7. The isolated anti-Cathepsin-D antibody of claim 6, wherein said antibody is able to activate and trigger the recruitment of natural killer cells into tumors and to induce cytotoxicity.
8. The isolated anti-Cathepsin-D antibody of any one of claims 1 to 4, wherein said antibody comprises a Fc region comprising at least 3 mutations selected from the group consisting of L234A, L235A and P329G (“F1M1-Fc-”).
9. The isolated anti-Cathepsin-D antibody of any one of claims 1 to 8, wherein the anti- Cathepsin-D antibody is a human antibody.
10. The isolated anti-Cathepsin-D antibody of any one of claims 1 to 9, wherein said antibody inhibits the tumor recruitment of immunosuppressive tumor-associated macrophages (TAMs) M2, is able to enhance the activation of anti-tumor M1-polarized TAMs, is able to reduce the expression of exhaustion markers on CD4+ and CD8+ T cells, such as PD-L1 and LAG3, in tumors and draining lymph nodes, and to promote the recruitment and maturation of conventional cDC1 dendritic cells.
11. The isolated anti-Cathepsin-D antibody of any one of claim 1 to 10, wherein said antibody is conjugated to a therapeutic moiety, and in particularly a cytotoxic moiety.
12. A cross-competing antibody which cross-competes for binding Cathepsin-D with the antibody according to any one of claims 1 to 11.
13. A nucleic acid molecule encoding the anti-Cathepsin-D antibody of any one of claims 1 to 12.
14. A vector comprising the nucleic acid molecule of claim 13.
15. A host cell which has been transfected, infected or transformed by the nucleic acid of claim 13 and/or the vector of claim 14.
16. A pharmaceutical composition comprising the anti-Cathepsin-D antibody of any one of claims 1 to 12.
17. The human isolated human anti-Cathepsin-D antibody of any one of claim 1 to 12 or the pharmaceutical composition of claim 16 for use as a drug.
18. A method for treating hyperproliferative disorders or diseases in a subject in need thereof, comprising administering to said subject an effective amount of the anti- Cathepsin-D antibody of any one of claim 1 to 12 or the pharmaceutical composition of claim 16.
19. The method for treating hyperproliferative disorders or diseases of claim 18, wherein the hyperproliferative disease is cancer and more particularly a cancer selected from the group consisting of breast cancer, melanoma, ovarian cancer, lung cancer, liver cancer, pancreatic cancer, melanoma, squamous cell carcinoma, endometrial cancer, head and neck cancer, bladder cancer, malignant glioma, prostate cancer, colon adenocarcinoma or gastric cancer.
20. The method for treating hyperproliferative disorders or diseases of claim 18, wherein the cancer is triple-negative breast cancer (TNBC).
21. The method for treating cancer of any one of claim 18 or 19, wherein said anti- Cathepsin-D antibody is administered in combination with anti-cancer therapy selected from the groups consisting of radiation therapy, immune checkpoint inhibitor, antiandrogens, CAR therapy such as T-, M- or CAR NK-cell therapy, antibody-drug conjugates (ADC) or chemotherapeutic agent 22. The method for treating cancer of claim 19, wherein the chemotherapeutic agent is paclitaxel. 23. The method for treating cancer of claim 19, wherein the antiandrogen is enzalutamide.
PCT/EP2024/0636252023-05-172024-05-16Anti-cathepsin-d antibodiesWO2024236156A1 (en)

Applications Claiming Priority (2)

Application NumberPriority DateFiling DateTitle
EP233057892023-05-17
EP23305789.22023-05-17

Publications (1)

Publication NumberPublication Date
WO2024236156A1true WO2024236156A1 (en)2024-11-21

Family

ID=86760327

Family Applications (1)

Application NumberTitlePriority DateFiling Date
PCT/EP2024/063625WO2024236156A1 (en)2023-05-172024-05-16Anti-cathepsin-d antibodies

Country Status (1)

CountryLink
WO (1)WO2024236156A1 (en)

Citations (82)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US244A (en)1837-06-30Edward flint
US5204A (en)1847-07-24james cantelo
US4486414A (en)1983-03-211984-12-04Arizona Board Of ReagentsDolastatins A and B cell growth inhibitory substances
EP0154316A2 (en)1984-03-061985-09-11Takeda Chemical Industries, Ltd.Chemically modified lymphokine and production thereof
EP0173494A2 (en)1984-08-271986-03-05The Board Of Trustees Of The Leland Stanford Junior UniversityChimeric receptors by DNA splicing and expression
WO1987002671A1 (en)1985-11-011987-05-07International Genetic Engineering, Inc.Modular assembly of antibody genes, antibodies prepared thereby and use
US4816567A (en)1983-04-081989-03-28Genentech, Inc.Recombinant immunoglobin preparations
US4816444A (en)1987-07-101989-03-28Arizona Board Of Regents, Arizona State UniversityCell growth inhibitory substance
EP0322094A1 (en)1987-10-301989-06-28Delta Biotechnology LimitedN-terminal fragments of human serum albumin
US4861719A (en)1986-04-251989-08-29Fred Hutchinson Cancer Research CenterDNA constructs for retrovirus packaging cell lines
US4879278A (en)1989-05-161989-11-07Arizona Board Of RegentsIsolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15
US4880935A (en)1986-07-111989-11-14Icrf (Patents) LimitedHeterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
WO1989012624A2 (en)1988-06-141989-12-28Cetus CorporationCoupling agents and sterically hindered disulfide linked conjugates prepared therefrom
EP0401384A1 (en)1988-12-221990-12-12Kirin-Amgen, Inc.Chemically modified granulocyte colony stimulating factor
US4978744A (en)1989-01-271990-12-18Arizona Board Of RegentsSynthesis of dolastatin 10
US4986988A (en)1989-05-181991-01-22Arizona Board Of RegentsIsolation and structural elucidation of the cytostatic linear depsipeptides dolastatin 13 and dehydrodolastatin 13
US5076973A (en)1988-10-241991-12-31Arizona Board Of RegentsSynthesis of dolastatin 3
EP0486525A1 (en)1989-08-011992-05-27Cemu Bioteknik AbStabilized protein or peptide conjugates.
US5122368A (en)1988-02-111992-06-16Bristol-Myers Squibb CompanyAnthracycline conjugates having a novel linker and methods for their production
US5138036A (en)1989-11-131992-08-11Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityIsolation and structural elucidation of the cytostatic cyclodepsipeptide dolastatin 14
US5202238A (en)1987-10-271993-04-13OncogenProduction of chimeric antibodies by homologous recombination
US5278056A (en)1988-02-051994-01-11The Trustees Of Columbia University In The City Of New YorkRetroviral packaging cell lines and process of using same
WO1994019478A1 (en)1993-02-221994-09-01The Rockefeller UniversityProduction of high titer helper-free retroviruses by transient transfection
US5410024A (en)1993-01-211995-04-25Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide amides
WO1995014785A1 (en)1993-11-231995-06-01Rhone-Poulenc Rorer S.A.Composition for the in vivo production of therapeutic products
US5476996A (en)1988-06-141995-12-19Lidak PharmaceuticalsHuman immune system in non-human animal
US5504191A (en)1994-08-011996-04-02Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide methyl esters
US5521284A (en)1994-08-011996-05-28Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide amides and esters
US5530097A (en)1994-08-011996-06-25Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory peptide amides
WO1996022378A1 (en)1995-01-201996-07-25Rhone-Poulenc Rorer S.A.Cells for the production of recombinant adenoviruses
US5554725A (en)1994-09-141996-09-10Arizona Board Of Regents Acting On Behalf Of Arizona State UniversitySynthesis of dolastatin 15
US5585089A (en)1988-12-281996-12-17Protein Design Labs, Inc.Humanized immunoglobulins
US5599902A (en)1994-11-101997-02-04Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityCancer inhibitory peptides
US5622929A (en)1992-01-231997-04-22Bristol-Myers Squibb CompanyThioether conjugates
US5624821A (en)1987-03-181997-04-29Scotgen Biopharmaceuticals IncorporatedAntibodies with altered effector functions
US5635483A (en)1992-12-031997-06-03Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityTumor inhibiting tetrapeptide bearing modified phenethyl amides
US5663149A (en)1994-12-131997-09-02Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US5677425A (en)1987-09-041997-10-14Celltech Therapeutics LimitedRecombinant antibody
US5714350A (en)1992-03-091998-02-03Protein Design Labs, Inc.Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5770429A (en)1990-08-291998-06-23Genpharm International, Inc.Transgenic non-human animals capable of producing heterologous antibodies
US5780588A (en)1993-01-261998-07-14Arizona Board Of RegentsElucidation and synthesis of selected pentapeptides
WO1998045322A2 (en)1997-04-101998-10-15Royal Netherlands Academy Of Arts And SciencesDiagnosis method and reagents
US5824805A (en)1995-12-221998-10-20King; DaltonBranched hydrazone linkers
US5859205A (en)1989-12-211999-01-12Celltech LimitedHumanised antibodies
US5869046A (en)1995-04-141999-02-09Genentech, Inc.Altered polypeptides with increased half-life
WO1999010494A2 (en)1997-08-251999-03-04Genentech, Inc.Agonist antibodies to the thrombopoietin receptor, and their therapeutic uses
US5882877A (en)1992-12-031999-03-16Genzyme CorporationAdenoviral vectors for gene therapy containing deletions in the adenoviral genome
US6013516A (en)1995-10-062000-01-11The Salk Institute For Biological StudiesVector and method of use for nucleic acid delivery to non-dividing cells
US6034065A (en)1992-12-032000-03-07Arizona Board Of RegentsElucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10
US6075181A (en)1990-01-122000-06-13Abgenix, Inc.Human antibodies derived from immunized xenomice
WO2000042072A2 (en)1999-01-152000-07-20Genentech, Inc.Polypeptide variants with altered effector function
US6121022A (en)1995-04-142000-09-19Genentech, Inc.Altered polypeptides with increased half-life
US6130237A (en)1996-09-122000-10-10Cancer Research Campaign Technology LimitedCondensed N-aclyindoles as antitumor agents
US6150584A (en)1990-01-122000-11-21Abgenix, Inc.Human antibodies derived from immunized xenomice
US6214345B1 (en)1993-05-142001-04-10Bristol-Myers Squibb Co.Lysosomal enzyme-cleavable antitumor drug conjugates
US6239104B1 (en)1997-02-252001-05-29Arizona Board Of RegentsIsolation and structural elucidation of the cytostatic linear and cyclo-depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
US6277375B1 (en)1997-03-032001-08-21Board Of Regents, The University Of Texas SystemImmunoglobulin-like domains with increased half-lives
US6323315B1 (en)1999-09-102001-11-27Basf AktiengesellschaftDolastatin peptides
WO2002088172A2 (en)2001-04-302002-11-07Seattle Genetics, Inc.Pentapeptide compounds and uses related thereto
US20030083263A1 (en)2001-04-302003-05-01Svetlana DoroninaPentapeptide compounds and uses related thereto
WO2003063792A2 (en)2002-01-302003-08-07The Brigham And Women's Hospital, Inc.Compositions and methods related to tim-3, a th1-specific cell surface molecule
US20030153043A1 (en)1997-05-212003-08-14Biovation LimitedMethod for the production of non-immunogenic proteins
WO2004010957A2 (en)2002-07-312004-02-05Seattle Genetics, Inc.Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US20050014934A1 (en)2002-10-152005-01-20Hinton Paul R.Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7041870B2 (en)2000-11-302006-05-09Medarex, Inc.Transgenic transchromosomal rodents for making human antibodies
US7189826B2 (en)1997-11-242007-03-13Institute For Human Genetics And BiochemistryMonoclonal human natural antibodies
US20070061900A1 (en)2000-10-312007-03-15Murphy Andrew JMethods of modifying eukaryotic cells
US7235641B2 (en)2003-12-222007-06-26Micromet AgBispecific antibodies
US7432059B2 (en)2000-06-282008-10-07Dana-Farber Cancer Institute, Inc.Methods of identifying compounds that upmodulate T cell activation in the presence of a PD-1 mediated signal
WO2009054864A1 (en)2007-10-262009-04-30Rigel Pharmaceuticals, Inc.Polycyclic aryl substituted triazoles and polycyclic heteroaryl substituted triazoles useful as axl inhibitors
WO2010106180A2 (en)2009-03-202010-09-23Lfb BiotechnologiesOptimized fc variants
WO2010117057A1 (en)2009-04-102010-10-14協和発酵キリン株式会社Method for treatment of blood tumor using anti-tim-3 antibody
WO2011155607A1 (en)2010-06-112011-12-15協和発酵キリン株式会社Anti-tim-3 antibody
WO2012059882A2 (en)2010-11-052012-05-10Rinat Neuroscience CorporationEngineered polypeptide conjugates and methods for making thereof using transglutaminase
WO2013006490A2 (en)2011-07-012013-01-10Cellerant Therapeutics, Inc.Antibodies that specifically bind to tim3
US8552154B2 (en)2008-09-262013-10-08Emory UniversityAnti-PD-L1 antibodies and uses therefor
WO2014150677A1 (en)2013-03-152014-09-25Bristol-Myers Squibb CompanyInhibitors of indoleamine 2,3-dioxygenase (ido)
WO2015004400A1 (en)2013-07-112015-01-15Universite Francois RabelaisNovel antibody-drug conjugates and the use of same in therapy
WO2015117002A1 (en)2014-01-312015-08-06Novartis AgAntibody molecules to tim-3 and uses thereof
WO2016145536A1 (en)*2015-03-182016-09-22Immunobiochem CorporationConjugates for the treatment of cancer targeted at intracellular tumor-associated antigens
WO2016188911A1 (en)2015-05-222016-12-01INSERM (Institut National de la Santé et de la Recherche Médicale)Human monoclonal antibodies fragments inhibiting both the cath-d catalytic activity and its binding to the lrp1 receptor
WO2020127411A1 (en)2018-12-192020-06-25INSERM (Institut National de la Santé et de la Recherche Médicale)Methods and compositions for treating cancers by immuno-modulation using antibodies against cathespin-d

Patent Citations (89)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US5204A (en)1847-07-24james cantelo
US244A (en)1837-06-30Edward flint
US4486414A (en)1983-03-211984-12-04Arizona Board Of ReagentsDolastatins A and B cell growth inhibitory substances
US4816567A (en)1983-04-081989-03-28Genentech, Inc.Recombinant immunoglobin preparations
EP0154316A2 (en)1984-03-061985-09-11Takeda Chemical Industries, Ltd.Chemically modified lymphokine and production thereof
EP0173494A2 (en)1984-08-271986-03-05The Board Of Trustees Of The Leland Stanford Junior UniversityChimeric receptors by DNA splicing and expression
WO1987002671A1 (en)1985-11-011987-05-07International Genetic Engineering, Inc.Modular assembly of antibody genes, antibodies prepared thereby and use
US4861719A (en)1986-04-251989-08-29Fred Hutchinson Cancer Research CenterDNA constructs for retrovirus packaging cell lines
US4880935A (en)1986-07-111989-11-14Icrf (Patents) LimitedHeterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
US5624821A (en)1987-03-181997-04-29Scotgen Biopharmaceuticals IncorporatedAntibodies with altered effector functions
US5648260A (en)1987-03-181997-07-15Scotgen Biopharmaceuticals IncorporatedDNA encoding antibodies with altered effector functions
US4816444A (en)1987-07-101989-03-28Arizona Board Of Regents, Arizona State UniversityCell growth inhibitory substance
US5677425A (en)1987-09-041997-10-14Celltech Therapeutics LimitedRecombinant antibody
US5202238A (en)1987-10-271993-04-13OncogenProduction of chimeric antibodies by homologous recombination
EP0322094A1 (en)1987-10-301989-06-28Delta Biotechnology LimitedN-terminal fragments of human serum albumin
US5278056A (en)1988-02-051994-01-11The Trustees Of Columbia University In The City Of New YorkRetroviral packaging cell lines and process of using same
US5122368A (en)1988-02-111992-06-16Bristol-Myers Squibb CompanyAnthracycline conjugates having a novel linker and methods for their production
US5698767A (en)1988-06-141997-12-16Lidak PharmaceuticalsHuman immune system in non-human animal
US5476996A (en)1988-06-141995-12-19Lidak PharmaceuticalsHuman immune system in non-human animal
WO1989012624A2 (en)1988-06-141989-12-28Cetus CorporationCoupling agents and sterically hindered disulfide linked conjugates prepared therefrom
US5076973A (en)1988-10-241991-12-31Arizona Board Of RegentsSynthesis of dolastatin 3
EP0401384A1 (en)1988-12-221990-12-12Kirin-Amgen, Inc.Chemically modified granulocyte colony stimulating factor
US5585089A (en)1988-12-281996-12-17Protein Design Labs, Inc.Humanized immunoglobulins
US4978744A (en)1989-01-271990-12-18Arizona Board Of RegentsSynthesis of dolastatin 10
US4879278A (en)1989-05-161989-11-07Arizona Board Of RegentsIsolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15
US4986988A (en)1989-05-181991-01-22Arizona Board Of RegentsIsolation and structural elucidation of the cytostatic linear depsipeptides dolastatin 13 and dehydrodolastatin 13
EP0486525A1 (en)1989-08-011992-05-27Cemu Bioteknik AbStabilized protein or peptide conjugates.
US5138036A (en)1989-11-131992-08-11Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityIsolation and structural elucidation of the cytostatic cyclodepsipeptide dolastatin 14
US5859205A (en)1989-12-211999-01-12Celltech LimitedHumanised antibodies
US6075181A (en)1990-01-122000-06-13Abgenix, Inc.Human antibodies derived from immunized xenomice
US6150584A (en)1990-01-122000-11-21Abgenix, Inc.Human antibodies derived from immunized xenomice
US5770429A (en)1990-08-291998-06-23Genpharm International, Inc.Transgenic non-human animals capable of producing heterologous antibodies
US5622929A (en)1992-01-231997-04-22Bristol-Myers Squibb CompanyThioether conjugates
US5714350A (en)1992-03-091998-02-03Protein Design Labs, Inc.Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US6350861B1 (en)1992-03-092002-02-26Protein Design Labs, Inc.Antibodies with increased binding affinity
US6034065A (en)1992-12-032000-03-07Arizona Board Of RegentsElucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10
US5882877A (en)1992-12-031999-03-16Genzyme CorporationAdenoviral vectors for gene therapy containing deletions in the adenoviral genome
US5635483A (en)1992-12-031997-06-03Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityTumor inhibiting tetrapeptide bearing modified phenethyl amides
US5410024A (en)1993-01-211995-04-25Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide amides
US5780588A (en)1993-01-261998-07-14Arizona Board Of RegentsElucidation and synthesis of selected pentapeptides
WO1994019478A1 (en)1993-02-221994-09-01The Rockefeller UniversityProduction of high titer helper-free retroviruses by transient transfection
US6214345B1 (en)1993-05-142001-04-10Bristol-Myers Squibb Co.Lysosomal enzyme-cleavable antitumor drug conjugates
WO1995014785A1 (en)1993-11-231995-06-01Rhone-Poulenc Rorer S.A.Composition for the in vivo production of therapeutic products
US5665860A (en)1994-08-011997-09-09Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory peptide amides
US5530097A (en)1994-08-011996-06-25Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory peptide amides
US5521284A (en)1994-08-011996-05-28Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide amides and esters
US5504191A (en)1994-08-011996-04-02Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide methyl esters
US5554725A (en)1994-09-141996-09-10Arizona Board Of Regents Acting On Behalf Of Arizona State UniversitySynthesis of dolastatin 15
US5599902A (en)1994-11-101997-02-04Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityCancer inhibitory peptides
US5663149A (en)1994-12-131997-09-02Arizona Board Of Regents Acting On Behalf Of Arizona State UniversityHuman cancer inhibitory pentapeptide heterocyclic and halophenyl amides
WO1996022378A1 (en)1995-01-201996-07-25Rhone-Poulenc Rorer S.A.Cells for the production of recombinant adenoviruses
US6121022A (en)1995-04-142000-09-19Genentech, Inc.Altered polypeptides with increased half-life
US5869046A (en)1995-04-141999-02-09Genentech, Inc.Altered polypeptides with increased half-life
US6013516A (en)1995-10-062000-01-11The Salk Institute For Biological StudiesVector and method of use for nucleic acid delivery to non-dividing cells
US5824805A (en)1995-12-221998-10-20King; DaltonBranched hydrazone linkers
US6130237A (en)1996-09-122000-10-10Cancer Research Campaign Technology LimitedCondensed N-aclyindoles as antitumor agents
US6239104B1 (en)1997-02-252001-05-29Arizona Board Of RegentsIsolation and structural elucidation of the cytostatic linear and cyclo-depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
US6277375B1 (en)1997-03-032001-08-21Board Of Regents, The University Of Texas SystemImmunoglobulin-like domains with increased half-lives
WO1998045322A2 (en)1997-04-101998-10-15Royal Netherlands Academy Of Arts And SciencesDiagnosis method and reagents
US20030153043A1 (en)1997-05-212003-08-14Biovation LimitedMethod for the production of non-immunogenic proteins
WO1999010494A2 (en)1997-08-251999-03-04Genentech, Inc.Agonist antibodies to the thrombopoietin receptor, and their therapeutic uses
US7189826B2 (en)1997-11-242007-03-13Institute For Human Genetics And BiochemistryMonoclonal human natural antibodies
WO2000042072A2 (en)1999-01-152000-07-20Genentech, Inc.Polypeptide variants with altered effector function
US7371826B2 (en)1999-01-152008-05-13Genentech, Inc.Polypeptide variants with altered effector function
US6323315B1 (en)1999-09-102001-11-27Basf AktiengesellschaftDolastatin peptides
US7709214B2 (en)2000-06-282010-05-04Dana-Farber Cancer Institute, Inc.Methods for upregulating an immune response with agents that inhibit the intereaction between PD-L2 and PD-1
US7432059B2 (en)2000-06-282008-10-07Dana-Farber Cancer Institute, Inc.Methods of identifying compounds that upmodulate T cell activation in the presence of a PD-1 mediated signal
US20070061900A1 (en)2000-10-312007-03-15Murphy Andrew JMethods of modifying eukaryotic cells
US7041870B2 (en)2000-11-302006-05-09Medarex, Inc.Transgenic transchromosomal rodents for making human antibodies
US20030083263A1 (en)2001-04-302003-05-01Svetlana DoroninaPentapeptide compounds and uses related thereto
US6884869B2 (en)2001-04-302005-04-26Seattle Genetics, Inc.Pentapeptide compounds and uses related thereto
WO2002088172A2 (en)2001-04-302002-11-07Seattle Genetics, Inc.Pentapeptide compounds and uses related thereto
WO2003063792A2 (en)2002-01-302003-08-07The Brigham And Women's Hospital, Inc.Compositions and methods related to tim-3, a th1-specific cell surface molecule
WO2004010957A2 (en)2002-07-312004-02-05Seattle Genetics, Inc.Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US20050014934A1 (en)2002-10-152005-01-20Hinton Paul R.Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7235641B2 (en)2003-12-222007-06-26Micromet AgBispecific antibodies
WO2009054864A1 (en)2007-10-262009-04-30Rigel Pharmaceuticals, Inc.Polycyclic aryl substituted triazoles and polycyclic heteroaryl substituted triazoles useful as axl inhibitors
US8552154B2 (en)2008-09-262013-10-08Emory UniversityAnti-PD-L1 antibodies and uses therefor
WO2010106180A2 (en)2009-03-202010-09-23Lfb BiotechnologiesOptimized fc variants
WO2010117057A1 (en)2009-04-102010-10-14協和発酵キリン株式会社Method for treatment of blood tumor using anti-tim-3 antibody
WO2011155607A1 (en)2010-06-112011-12-15協和発酵キリン株式会社Anti-tim-3 antibody
WO2012059882A2 (en)2010-11-052012-05-10Rinat Neuroscience CorporationEngineered polypeptide conjugates and methods for making thereof using transglutaminase
WO2013006490A2 (en)2011-07-012013-01-10Cellerant Therapeutics, Inc.Antibodies that specifically bind to tim3
WO2014150677A1 (en)2013-03-152014-09-25Bristol-Myers Squibb CompanyInhibitors of indoleamine 2,3-dioxygenase (ido)
WO2015004400A1 (en)2013-07-112015-01-15Universite Francois RabelaisNovel antibody-drug conjugates and the use of same in therapy
WO2015117002A1 (en)2014-01-312015-08-06Novartis AgAntibody molecules to tim-3 and uses thereof
WO2016145536A1 (en)*2015-03-182016-09-22Immunobiochem CorporationConjugates for the treatment of cancer targeted at intracellular tumor-associated antigens
WO2016188911A1 (en)2015-05-222016-12-01INSERM (Institut National de la Santé et de la Recherche Médicale)Human monoclonal antibodies fragments inhibiting both the cath-d catalytic activity and its binding to the lrp1 receptor
WO2020127411A1 (en)2018-12-192020-06-25INSERM (Institut National de la Santé et de la Recherche Médicale)Methods and compositions for treating cancers by immuno-modulation using antibodies against cathespin-d

Non-Patent Citations (70)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. NP 001900
"Monoclonal Antibodies For Cancer Detection And Therapy", 1985, ACADEMIC PRESS, article "Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody In Cancer Therapy"
AGNEW CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
ARNON ET AL.: "Monoclonal Antibodies And Cancer Therapy", 1985, ALAN R. LISS, INC., article "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy"
ASHRAF YMANSOURI HLAURENT-MATHA VALCARAZ LBROGER PGUIU S ET AL.: "Immunotherapy of triple-negative breast cancer with cathepsin D-targeting antibodies", J IMMUNOTHER CANCER, vol. 7, no. 1, 2019, pages 29, XP021269907, DOI: 10.1186/s40425-019-0498-z
AXUP, J.Y., BAJJURI, K.M., RITLAND, M., HUTCHINS, B.M., KIM, C.H., KAZANE, S.A., HALDER, R., FORSYTH, J.S., SANTIDRIAN, A.F., STAF: "Synthesis of site-specific antibody-drug conjugates using unnatural amino acids", PROC. NATL. ACAD. SCI. USA, vol. 109, 2012, pages 16101 - 16106, XP055076259, DOI: 10.1073/pnas.1211023109
BARROSO-SOUSA RJAIN ECOHEN OKIM DBUENDIA-BUENDIA JWINER E ET AL.: "Prevalence and mutational determinants of high tumor mutation burden in breast cancer", ANN ONCOL, vol. 31, no. 3, 2020, pages 387 - 94
BIANCHINI GDE ANGELIS CLICATA LGIANNI L: "Treatment landscape of triple-negative breast cancer - expanded options, evolving needs", NAT REV CLIN ONCOL, vol. 19, no. 2, 2022, pages 91 - 113, XP037675445, DOI: 10.1038/s41571-021-00565-2
BOERNER ET AL., J. IMMUNOL., vol. 147, 1991, pages 86
CHARI ET AL., CANCER RES, vol. 52, 1992, pages 127 - 131
CONILH LSADILKOVA LVIRICEL WDUMONTET C: "Payload diversification: a key step in the development of antibody-drug conjugates", J HEMATOL ONCOL, vol. 16, no. 1, 17 January 2023 (2023-01-17), pages 3
CORREIA ALGUIMARAES JCAUF DER MAUR P ET AL.: "Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy", NATURE, vol. 594, no. 7864, 2021, pages 566 - 571, XP037617186, DOI: 10.1038/s41586-021-03614-z
CORTES JRUGO HSCESCON DWIM SAYUSOF MMGALLARDO C ET AL.: "Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer", N ENGL J MED, vol. 387, no. 3, 2022, pages 217 - 26, XP093162423, DOI: 10.1056/NEJMoa2202809
CURTIS ET AL., BRITISH JOURNAL OF PHARMACOLOGY ON EXPERIMENTAL DESIGN AND ANALYSIS IN PHARMACOLOGY, 2018
DAVID TIMOTHÉE ET AL: "Anti-cathepsin D immunotherapy triggers both innate and adaptive anti-tumour immunity in breast cancer", BRITISH JOURNAL OF PHARMACOLOGY, 26 January 2024 (2024-01-26), UK, XP093187298, ISSN: 0007-1188, DOI: 10.1111/bph.16291*
DENKERT CVON MINCKWITZ GDARB-ESFAHANI SLEDERER BHEPPNER BIWEBER KE ET AL.: "Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy", LANCET ONCOL, 2017
DESROYS DU ROURE PÉNÉLOPE ET AL: "A novel Fc-engineered cathepsin D-targeting antibody enhances ADCC, triggers tumor-infiltrating NK cell recruitment, and improves treatment with paclitaxel and enzalutamide in triple-negative breast cancer", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 12, no. 1, 30 January 2024 (2024-01-30), GB, pages e007135, XP093187293, ISSN: 2051-1426, Retrieved from the Internet <URL:https://jitc.bmj.com/content/jitc/12/1/e007135.full.pdf> DOI: 10.1136/jitc-2023-007135*
DRAGOVICH PS: "Degrader-antibody conjugates", CHEM SOC REV., vol. 51, no. 10, 23 May 2022 (2022-05-23), pages 3886 - 3897, XP093013565, DOI: 10.1039/D2CS00141A
DUBOWCHIKWALKER, PHARM. THERAPEUTICS, vol. 83, 1999, pages 67 - 123
FUXE ET AL., SEMIN CANCER BIOL, vol. 22, 2012, pages 455 - 461
GEORGOUDAKI ET AL., CELL REPORTS, vol. 15, 2016, pages 2000 - 11
GUYER ET AL., J. IMMUNOL., vol. 117, 1976, pages 587
HARLOW ET AL.: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY PRESS
HELLSTROM ET AL.: "Monoclonal Antibody Production Techniques and Applications", 1987, MARCEL DEIKER, INC., article "Antibodies For Drug Delivery", pages: 51 - 63
HOOGENBOOM ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581
JIANG KDONG MLI CSHENG J: "Unraveling Heterogeneity of Tumor Cells and Microenvironment and Its Clinical Implications for Triple Negative Breast Cancer", FRONT ONCOL, vol. 11, 2021, pages 557477
JOHNSON ET AL., ANTICANCER RES, vol. 15, 1995, pages 1387 - 93
JUNUTULA, J.R.FLAGELLA, K.M.GRAHAM, R.A.PARSONS, K.L.HA, E.RAAB, H.BHAKTA, S.NGUYEN, T.DUGGER, D.L.LI, G. ET AL.: "Engineered thio-trastuzumab-DM1 conjugate with an improved therapeutic index to target humanepidermal growth factor receptor 2-positive breast cancer", CLIN. CANCER RES., vol. 16, 2010, pages 4769 - 4778, XP002727053, DOI: 10.1158/1078-0432.CCR-10-0987
KANG JYU YJEONG SLEE HHEO HJPARK JJ ET AL.: "Prognostic role of high cathepsin D expression in breast cancer: a systematic review and meta-analysis", THER ADV MED ONCOL, vol. 12, 2020, pages 1758835920927838
KIM ET AL., J. IMMUNOL., vol. 24, 1994, pages 249
KOZBOR, J. IMMUNOL., vol. 13, 1984, pages 3001
KYRIAZOGLOU AKAPARELOU MGOUMAS GLIONTOS MZAKOPOULOU RZOGRAFOS E ET AL.: "Immunotherapy in HER2-Positive Breast Cancer: A Systematic Review", BREAST CARE (BASEL, vol. 17, no. 1, 2022, pages 63 - 70
LAU ET AL., BIOORG-MED-CHEM., vol. 3, no. 10, 1995, pages 1305 - 1304
LEFRANC M.-P.: "The IMGT unique numbering for Immunoglobulins, T cell receptors and Ig-like domains", THE IMMUNOLOGIST, vol. 7, 1999, pages 132 - 136
LEFRANC M.-P.: "Unique database numbering system for immunogenetic analysis", IMMUNOLOGY TODAY, vol. 18, 1997, pages 509, XP004093509, DOI: 10.1016/S0167-5699(97)01163-8
LEFRANC, M.-P.POMMIÉ, C.RUIZ, M.GIUDICELLI, V.FOULQUIER, E.TRUONG, L.THOUVENIN-CONTET, V.LEFRANC, G.: "IMGT unique numbering for immunoglobulin and T cell receptor variable domains and Ig superfamily V-like domains", DEV. COMP. IMMUNOL., vol. 27, 2003, pages 55 - 77, XP055585227, DOI: 10.1016/S0145-305X(02)00039-3
LI ET AL., CANCER RES, vol. 42, 1982, pages 999 - 1004
LI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 3557 - 3562
LIU ROLDHAM RJTEAL E ET AL.: "Fc-Engineering for Modulated Effector Functions-Improving Antibodies for Cancer Treatment", ANTIBODIES (BASEL, vol. 9, no. 4, 2020, XP055918764, DOI: 10.3390/antib9040064
LONBERG, CUR. OPIN.IMMUNOL., vol. 20, 2008, pages 450 - 459
LONBERG, NAT.BIOTECH., vol. 23, 2005, pages 1117 - 1125
LOPEZ-SOTO AGONZALEZ SSMYTH MJ ET AL.: "Control of Metastasis by NK Cells", CANCER CELL, vol. 32, no. 2, 2017, pages 135 - 154, XP055424288, DOI: 10.1016/j.ccell.2017.06.009
MANSOURI HALCARAZ LBMOLLEVI CMALLAVIALLE AJACOT WBOISSIERE-MICHOT F ET AL.: "Co-Expression of Androgen Receptor and Cathepsin D Defines a Triple-Negative Breast Cancer Subgroup with Poorer Overall Survival", CANCERS (BASEL, vol. 12, no. 5, 2020
MELLMAN ET AL., NATURE, vol. 480, 2011, pages 480 - 489
MULLER: "Meth. Enzymol.", vol. 92, 1983, pages: 589 - 601
NEVILLE ET AL., BIOL. CHEM., vol. 264, 1989, pages 14653 - 14661
NI, XIANDAI MIANYIXUE, vol. 26, no. 4, 2006, pages 265 - 268
PARDOLL, NATURE REV CANCER, vol. 12, 2012, pages 252 - 264
PRAGER IWATZL C: "Mechanisms of natural killer cell-mediated cellular cytotoxicity", J LEUKOC BIOL, vol. 105, no. 6, 2019, pages 1319 - 1329
RUGO HSIM SACARDOSO F ET AL.: "Efficacy of Margetuximab vs Trastuzumab in Patients With Pretreated ERBB2-Positive Advanced Breast Cancer: A Phase 3 Randomized Clinical Trial", JAMA ONCOL, vol. 7, no. 4, 2021, pages 573 - 584
SCOTT AMWOLCHOK JDOLD LJ: "Antibody therapy of cancer", NAT REV CANCER, vol. 12, no. 4, 2012, pages 278 - 87, XP055531673, DOI: 10.1038/nrc3236
SHIELDS, R. L. ET AL., J. BIOL. CHEN., vol. 276, 2001, pages 6591 - 6604
SOUZA-FONSECA-GUIMARAES F: "NK cell-based immunotherapies: awakening the innate anti-cancer response", DISCOV MED, vol. 21, no. 115, 2016, pages 197 - 203
TARIQ MZHANG JLIANG GDING LHE QYANG B: "Macrophage Polarization: Anti-Cancer Strategies to Target Tumor-Associated Macrophage in Breast Cancer", J CELL BIOCHEM, vol. 118, no. 9, 2017, pages 2484 - 501
THORPE ET AL., CANCER RES., vol. 47, 1987, pages 5924 - 5931
THORPE ET AL., IMMUNOL. REV., vol. 62, 1982, pages 119 - 58
THORPE ET AL.: "Monoclonal Antibodies '84: Biological And Clinical Applications", 1985, article "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review"
TRINCHIERI GVALIANTE N: "Receptors for the Fc fragment of IgG on natural killer cells", NAT IMMUN, vol. 12, no. 4-5, 1993, pages 218 - 34
VAN DIJKVAN DE WINKEL, CUR. OPIN. PHARMACOL., vol. 5, 2001, pages 368 - 74
VIGNON FCAPONY FCHAMBON MFREISS GGARCIA MROCHEFORT H: "Autocrine growth stimulation of the MCF 7 breast cancer cells by the estrogen-regulated 52 K protein", ENDOCRINOLOGY, vol. 118, no. 4, 1986, pages 1537 - 45
VOLLMERSBRANDLEIN, HISTOLOGY AND HISTOPATHOLOGY, vol. 20, no. 3, 2005, pages 927 - 937
VOLLMERSBRANDLEIN, METHODS AND FINDINGS IN EXPERIMENTAL AND CLINICAL PHARMACOLOGY, vol. 27, no. 3, 2005, pages 185 - 91
VOSKOBOINIK IWHISSTOCK JCTRAPANI JA: "Perforin and granzymes: function, dysfunction and human pathology", NAT REV IMMUNOL, vol. 15, no. 6, 2015, pages 388 - 400
WANG SYANG YMA PZHA YZHANG JLEI ALI N: "CAR-macrophage: An extensive immune enhancer to fight cancer", EBIOMEDICINE, vol. 76, February 2022 (2022-02-01), pages 103873, XP055964896, DOI: 10.1016/j.ebiom.2022.103873
WARREN HSKINNEAR BF: "Quantitative analysis of the effect of CD16 ligation on human NK cell proliferation", J IMMUNOL., vol. 162, no. 2, 1999, pages 735 - 42
WAWRZYNCZAK ET AL.: "mmunoconjugates: Antibody Conjugates in Radioimagery and Therapy of Cancer", 1987, OXFORD U. PRESS
WOJTUKIEWICZ MZPOGORZELSKA MPOLITYNSKA B: "Immunotherapy for triple negative breast cancer: the end of the beginning or the beginning of the end?", CANCER METASTASIS REV, vol. 41, no. 3, 2022, pages 465 - 9
YAHYA ASHRAF ET AL: "Immunotherapy of triple-negative breast cancer with cathepsin D-targeting antibodies", JOURNAL FOR IMMUNOTHERAPY OF CANCER, BIOMED CENTRAL LTD, LONDON, UK, vol. 7, no. 1, 4 February 2019 (2019-02-04), pages 1 - 17, XP021269907, DOI: 10.1186/S40425-019-0498-Z*
ZHAO ET AL., ONCOTARGET, vol. 8, 2017, pages 30576 - 86
ZHAO ET AL.: "Molecular subtypes and precision treatment of triple-negative breast cancer", ANN TRANSL MED, vol. 8, no. 7, April 2020 (2020-04-01), pages 499

Similar Documents

PublicationPublication DateTitle
JP7611196B2 (en) Diagnosis and treatment of cancer involving cancer stem cells
US20230303696A1 (en)Bispecific Anti-PSMA x Anti-CD28 Antibodies and Uses Thereof
US20230052212A1 (en)Fgfr2 inhibitors alone or in combination with immune stimulating agents in cancer treatment
US11274160B2 (en)Antibodies having specificity to Nectin-4 and uses thereof
RU2765410C2 (en)Methods for treating cancer, including tigit-binding agents
AU2014302812A1 (en)Use of semaphorin-4D inhibitory molecules in combination with an immune modulating therapy to inhibit tumor growth and metastases
JP2016534090A (en) Medical use of CD38 agonist
JP2018524300A (en) Methods and antibodies for modulating immune responses
JP2023129433A (en)NOVEL ANTIBODIES AND COMBINED USE OF Treg-DEPLETING ANTIBODY AND IMMUNOSTIMULATORY ANTIBODY
KR20220035150A (en) Antibody combinations for the treatment of cancer in certain patients
WO2023034963A2 (en)Combinations of immunotherapies and uses thereof
US20210196744A1 (en)Compositions for cancer therapy and methods
WO2020109627A1 (en)Anti-neuropilin-1 and anti-programmed cell death-1 combination therapy for treating cancer
US11186634B2 (en)Antibodies targeting tumor associated macrophages and uses thereof
TW202340243A (en)Anti-cd39 antibodies and use thereof
US20230181753A1 (en)New method to treat cutaneous t-cell lymphomas and tfh derived lymphomas
WO2024236156A1 (en)Anti-cathepsin-d antibodies
JP2022553643A (en) Compositions and methods for treating cytotoxic T-cell resistant tumors
CN113905747A (en) Cells, compositions and methods for enhancing immune function
US20230040928A1 (en)Antibodies having specificity to her4 and uses thereof
WO2023056361A1 (en)Anti-hsp70 antibodies and therapeutic uses thereof
KR20250005988A (en) KIR3DL3 inhibitor and immune cell activator
KR20250059415A (en) B7-H3 antigen-binding molecule
CN119053340A (en)Multispecific binding agents directed against PD-L1 and CD137 in combination with anti-PD-1 antibodies for the treatment of cancer
CN118742323A (en) KIR3DL3 inhibitors and immune cell activators

Legal Events

DateCodeTitleDescription
121Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number:24727709

Country of ref document:EP

Kind code of ref document:A1


[8]ページ先頭

©2009-2025 Movatter.jp